1
|
Wang X, Chen S, Wang X, Song Z, Wang Z, Niu X, Chen X, Chen X. Application of artificial hibernation technology in acute brain injury. Neural Regen Res 2024; 19:1940-1946. [PMID: 38227519 DOI: 10.4103/1673-5374.390968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/20/2023] [Indexed: 01/17/2024] Open
Abstract
Controlling intracranial pressure, nerve cell regeneration, and microenvironment regulation are the key issues in reducing mortality and disability in acute brain injury. There is currently a lack of effective treatment methods. Hibernation has the characteristics of low temperature, low metabolism, and hibernation rhythm, as well as protective effects on the nervous, cardiovascular, and motor systems. Artificial hibernation technology is a new technology that can effectively treat acute brain injury by altering the body's metabolism, lowering the body's core temperature, and allowing the body to enter a state similar to hibernation. This review introduces artificial hibernation technology, including mild hypothermia treatment technology, central nervous system regulation technology, and artificial hibernation-inducer technology. Upon summarizing the relevant research on artificial hibernation technology in acute brain injury, the research results show that artificial hibernation technology has neuroprotective, anti-inflammatory, and oxidative stress-resistance effects, indicating that it has therapeutic significance in acute brain injury. Furthermore, artificial hibernation technology can alleviate the damage of ischemic stroke, traumatic brain injury, cerebral hemorrhage, cerebral infarction, and other diseases, providing new strategies for treating acute brain injury. However, artificial hibernation technology is currently in its infancy and has some complications, such as electrolyte imbalance and coagulation disorders, which limit its use. Further research is needed for its clinical application.
Collapse
Affiliation(s)
- Xiaoni Wang
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shulian Chen
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Xiaoyu Wang
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Zhen Song
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ziqi Wang
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaofei Niu
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaochu Chen
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Xuyi Chen
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| |
Collapse
|
2
|
Lv S, Geng X, Yun HJ, Ding Y. Phenothiazines reduced autophagy in ischemic stroke through endoplasmic reticulum (ER) stress-associated PERK-eIF2α pathway. Exp Neurol 2023; 369:114524. [PMID: 37673390 DOI: 10.1016/j.expneurol.2023.114524] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/15/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Neuroprotective effects have been the main focus of new treatment modalities for ischemic stroke. Phenothiazines, or chlorpromazine plus promethazine (C + P), are known to prevent the generation of free radicals and uptake of Ca2+ by plasma membrane; they have a potential as a treatment for acute ischemic stroke (AIS). This study aims to investigate the role of endoplasmic reticulum (ER) stress-associated PERK-eIF2α pathway underlying the phenothiazine-induced neuroprotective effects after cerebral ischemia/reperfusion (I/R) injury. METHODS A total of 49 male Sprague Dawley rats (280-320 g) were randomly divided into 4 groups (n = 7 per group): (1) sham, (2) I/R that received 2 h of middle cerebral artery occlusion (MCAO), followed by 6 or 24 h of reperfusion, (3) MCAO treated by C + P without temperature control and (4) MCAO treated by C + P with temperature control. Human neuroblastoma (SH-SY5Y) cells were used in 5 groups: (1) control, (2) oxygen-glucose deprivation (OGD) for 2 h followed by reoxygenation (OGD/R), (3) OGD/R with C + P; (4) OGD/R with PERK inhibitor, GSK2656157, and (5) OGD/R with C + P and GSK2656157. The molecules of ER stress, unfolded protein response (UPR) (Bip, PERK, p-PERK, p-PERK/PERK, eIF2α, p-eIF2α, p-eIF2α/eIF2α), autophagy (ATG12, LC3II/I), and apoptosis (BAX, Bcl-XL) were measured at mRNA levels by real time PCR and protein levels by Western blotting. RESULTS In ischemic rats followed by reperfusion, expression of Bip, p-PERK/PERK, p-eIF2α/eIF2α, ATG12, and LC3II/I, as well as BAX were all significantly increased. These markers were significantly reduced by C + P at both 6 and 24 h of reperfusion. Anti-apoptotic Bcl-XL expression was increased, while pro-apoptotic BAX expression was decreased by C + P. In SH-SY5Y cell lines, both C + P and GSK2656157 significantly reduced the level of autophagy and apoptosis after I/R, respectively. The combination of GSK2656157 and C + P did not promote the same effect, suggesting that C + P did not induce any neuroprotective effect by inhibiting autophagy and apoptosis through the PERK-eIF2α pathway when this pathway was already blocked by GSK2656157. In general, the reduction in body temperature by phenothiazines was associated with better neuroprotection but it did not reach significant levels. CONCLUSION The combined treatment of C + P plays a crucial role in stroke therapy by inhibiting ER stress-mediated autophagy, thereby leading to reduced apoptosis and increased neuroprotection. Our findings highlight the PERK-eIF2α pathway as a central mechanism through which C + P exerts its beneficial effects. The results from this study may pave the way for the development of more targeted and effective treatments for stroke patients.
Collapse
Affiliation(s)
- Shuyu Lv
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States.
| | - Ho Jun Yun
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
3
|
Wang R, Xiao L, Pan J, Bao G, Zhu Y, Zhu D, Wang J, Pei C, Ma Q, Fu X, Wang Z, Zhu M, Wang G, Gong L, Tong Q, Jiang M, Hu J, He M, Wang Y, Li T, Liang C, Li W, Xia C, Li Z, Ma DK, Tan M, Liu JY, Jiang W, Luo C, Yu B, Dang Y. Natural product P57 induces hypothermia through targeting pyridoxal kinase. Nat Commun 2023; 14:5984. [PMID: 37752106 PMCID: PMC10522591 DOI: 10.1038/s41467-023-41435-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
Induction of hypothermia during hibernation/torpor enables certain mammals to survive under extreme environmental conditions. However, pharmacological induction of hypothermia in most mammals remains a huge challenge. Here we show that a natural product P57 promptly induces hypothermia and decreases energy expenditure in mice. Mechanistically, P57 inhibits the kinase activity of pyridoxal kinase (PDXK), a key metabolic enzyme of vitamin B6 catalyzing phosphorylation of pyridoxal (PL), resulting in the accumulation of PL in hypothalamus to cause hypothermia. The hypothermia induced by P57 is significantly blunted in the mice with knockout of PDXK in the preoptic area (POA) of hypothalamus. We further found that P57 and PL have consistent effects on gene expression regulation in hypothalamus, and they may activate medial preoptic area (MPA) neurons in POA to induce hypothermia. Taken together, our findings demonstrate that P57 has a potential application in therapeutic hypothermia through regulation of vitamin B6 metabolism and PDXK serves as a previously unknown target of P57 in thermoregulation. In addition, P57 may serve as a chemical probe for exploring the neuron circuitry related to hypothermia state in mice.
Collapse
Affiliation(s)
- Ruina Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Xiao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Guangsen Bao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunmei Zhu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Di Zhu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chengfeng Pei
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Qinfeng Ma
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xian Fu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Ziruoyu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengdi Zhu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Guoxiang Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Gong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiuping Tong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Jiang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junchi Hu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tiejun Li
- Department of Pharmacology, College of Pharmacy, Naval Medical University, Shanghai, China
| | - Chunmin Liang
- Lab of Tumor Immunology, Department of Human Anatomy, Histology and Embryology, Basic Medical School of Fudan University, Shanghai, China
| | - Wei Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zengxia Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dengke K Ma
- Department of Physiology, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jun Yan Liu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Wei Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Biao Yu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| | - Yongjun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Xu S, Ji X, Li M, Wu D. Expedited brain cooling: Persistent temperature management from first aid to interhospital treatment. J Cereb Blood Flow Metab 2023; 43:319-321. [PMID: 36127836 PMCID: PMC9903226 DOI: 10.1177/0271678x221127088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 01/24/2023]
Abstract
Selective brain cooling is a promising technique for improving outcomes in ischemic stroke in the area of reperfusion. A recent study described the efficacy of a new method of selective brain cooling via active conductive head cooling. This is a major step forward in the administration of hypothermic treatment during pre-hospital transfer. However, to enhance the benefits of selective therapeutic cooling, a more comprehensive strategy preventing delay in hypothermic induction and increasing the accuracy of selectivity in the brain should be considered to mitigate the side effects related to therapeutic hypothermia.
Collapse
Affiliation(s)
- Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ming Li
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Chen X, An H, Wu D, Ji X. Research progress of selective brain cooling methods in the prehospital care for stroke patients: A narrative review. Brain Circ 2023; 9:16-20. [PMID: 37151794 PMCID: PMC10158655 DOI: 10.4103/bc.bc_88_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/14/2023] [Accepted: 01/29/2023] [Indexed: 05/09/2023] Open
Abstract
Over the past four decades, therapeutic hypothermia (TH) has long been suggested as a promising neuroprotective treatment of acute ischemic stroke (AIS). Much attention has focus on keeping the hypothermic benefits and removing side effects of systemic hypothermia. In the past few years, the advent of intravenous thrombolysis and endovascular thrombectomy has taken us into a reperfusion era of AIS treatment. With recent research emphasizing ways to plus neuroprotective treatments to reperfusion therapy, the spotlight is now shifting toward the study of how selective brain hypothermia can offset the drawbacks of systemic hypothermia and be applied in prehospital condition. This mini-review summarizes current brain cooling methods that can be used for inducing selective hypothermia in prehospital care. It will guide the future development of selective cooling methods, extend the application of TH in prehospital care, and provide insights into the prospects of selective hypothermia in AIS.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Hong An
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- Address for correspondence: Dr. Xunming Ji, Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China. E-mail:
| |
Collapse
|
6
|
Chlorpromazine and Promethazine (C+P) Reduce Brain Injury after Ischemic Stroke through the PKC-δ/NOX/MnSOD Pathway. Mediators Inflamm 2022; 2022:6886752. [PMID: 35873710 PMCID: PMC9307415 DOI: 10.1155/2022/6886752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/11/2022] [Indexed: 11/18/2022] Open
Abstract
Cerebral ischemia-reperfusion (I/R) incites neurologic damage through a myriad of complex pathophysiological mechanisms, most notably, inflammation and oxidative stress. In I/R injury, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) produces reactive oxygen species (ROS), which promote inflammatory and apoptotic pathways, augmenting ROS production and promoting cell death. Inhibiting ischemia-induced oxidative stress would be beneficial for reducing neuroinflammation and promoting neuronal cell survival. Studies have demonstrated that chlorpromazine and promethazine (C+P) induce neuroprotection. This study investigated how C+P minimizes oxidative stress triggered by ischemic injury. Adult male Sprague-Dawley rats were subject to middle cerebral artery occlusion (MCAO) and subsequent reperfusion. 8 mg/kg of C+P was injected into the rats when reperfusion was initiated. Neurologic damage was evaluated using infarct volumes, neurological deficit scoring, and TUNEL assays. NOX enzymatic activity, ROS production, protein expression of NOX subunits, manganese superoxide dismutase (MnSOD), and phosphorylation of PKC-δ were assessed. Neural SHSY5Y cells underwent oxygen-glucose deprivation (OGD) and subsequent reoxygenation and C+P treatment. We also evaluated ROS levels and NOX protein subunit expression, MnSOD, and p-PKC-δ/PKC-δ. Additionally, we measured PKC-δ membrane translocation and the level of interaction between NOX subunit (p47phox) and PKC-δ via coimmunoprecipitation. As hypothesized, treatment with C+P therapy decreased levels of neurologic damage. ROS production, NOX subunit expression, NOX activity, and p-PKC-δ/PKC-δ were all significantly decreased in subjects treated with C+P. C+P decreased membrane translocation of PKC-δ and lowered the level of interaction between p47phox and PKC-δ. This study suggests that C+P induces neuroprotective effects in ischemic stroke through inhibiting oxidative stress. Our findings also indicate that PKC-δ, NOX, and MnSOD are vital regulators of oxidative processes, suggesting that C+P may serve as an antioxidant.
Collapse
|
7
|
Li F, Gao J, Kohls W, Geng X, Ding Y. Perspectives on benefit of early and prereperfusion hypothermia by pharmacological approach in stroke. Brain Circ 2022; 8:69-75. [PMID: 35909706 PMCID: PMC9336590 DOI: 10.4103/bc.bc_27_22] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Stroke kills or disables approximately 15 million people worldwide each year. It is the leading cause of brain injury, resulting in persistent neurological deficits and profound physical handicaps. In spite of over 100 clinical trials, stroke treatment modalities are limited in applicability and efficacy, and therefore, identification of new therapeutic modalities is required to combat this growing problem. Poststroke oxidative damage and lactic acidosis are widely-recognized forms of brain ischemia/reperfusion injury. However, treatments directed at these injury mechanisms have not been effective. In this review, we offer a novel approach combining these well-established damage mechanisms with new insights into brain glucose handling. Specifically, emerging evidence of brain gluconeogenesis provides a missing link for understanding oxidative injury and lactate toxicity after ischemia. Therefore, dysfunctional gluconeogenesis may substantially contribute to oxidative and lactate damage. We further review that hypothermia initiated early in ischemia and before reperfusion may ameliorate gluconeogenic dysfunction and subsequently provide an important mechanism of hypothermic protection. We will focus on the efficacy of pharmacologically assisted hypothermia and suggest a combination that minimizes side effects. Together, this study will advance our knowledge of basic mechanisms of ischemic damage and apply this knowledge to develop new therapeutic strategies that are desperately needed in the clinical treatment of stroke.
Collapse
Affiliation(s)
- Fengwu Li
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Jie Gao
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Wesley Kohls
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Neurology, China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
8
|
Jiang Q, Wills M, Geng X, Ding Y. Chlorpromazine and promethazine reduces Brain injury through RIP1-RIP3 regulated activation of NLRP3 inflammasome following ischemic stroke. Neurol Res 2021; 43:668-676. [PMID: 33829970 DOI: 10.1080/01616412.2021.1910904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/27/2021] [Indexed: 12/26/2022]
Abstract
Objectives: Stroke is an important cause of death and disability. Recent evidence suggests that post-stroke inflammation is an important factor in stroke pathology and a root cause of its lasting consequences. Phenothiazine drugs, like chlorpromazine and promethazine (C + P), induce hypothermia and have been shown to play a major role in neuroprotection. In the present study, we investigated this neuroprotective mechanism by assessing the anti-inflammatory effect of these drugs.Methods: Adult Sprague-Dawley rats underwent 2 h of middle cerebral artery occlusion (MCAO) followed by 6 or 24 h of reperfusion, with or without C + P (8 mg/kg). Infarct volumes, neurological deficits, along with mRNA and protein quantities of receptor-interacting protein 1 (RIP1), receptor-interacting protein 3 (RIP3), NLRPyrin domain containing 3 (NLRP3), and interleukin-1β (IL-1β) were assessed, as well as the infiltration of neutrophils and macrophages.Results: C + P induced hypothermia that significantly reduced RIP1, RIP3, NLRP3 and IL-1β expression, infarction, and immune cell infiltration, while C + P treatment with temperature control at 37°C induced lesser effect.Conclusion: These findings suggest that the anti-inflammatory effect of C + P may be dependent on drug-induced hypothermia and regulation of the NLRP3 inflammasome via the RIP1/RIP3 complex. Future investigations are needed regarding C + P as potential treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Mélissa Wills
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
9
|
Zakharova NM, Tarahovsky YS, Komelina NP, Khrenov MO, Kovtun AL. Pharmacological torpor prolongs rat survival in lethal normobaric hypoxia. J Therm Biol 2021; 98:102906. [PMID: 34016333 DOI: 10.1016/j.jtherbio.2021.102906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
Resistance to hypoxia is one of the most prominent features of natural hibernation and is expected to be present in the pharmacological torpor (PT) that simulates hibernation. We studied resistance to lethal hypoxia (3.5% oxygen content) in rats under PT. To initiate PT, we used the previously developed pharmacological composition (PC) which, after a single intravenous injection, can induce a daily decrease in Tb by 7 °C-8 °C at the environmental temperature of 22 °C-23 °C. Half-survival (median) time of rats in lethal hypoxia was found to increase from 5 ± 0.8 min in anesthetized control rats to 150 ± 12 min in rats injected with PC, which is a 30-fold increase. Behavioral tests after PT and hypoxia, including the traveling distance, the number of rearing and grooming episodes, revealed that animal responses are significantly restored within a week. It is assumed that the discovered unprecedented resistance of artificially torpid rats to lethal hypoxia may open up broad prospects for the therapeutic use of PT for preconditioning to various damaging factors, treatment of diseases, and extend the so-called "golden hour" for lifesaving interventions.
Collapse
Affiliation(s)
| | - Yury S Tarahovsky
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia; Institute of Theoretical and Experimental Biophysics, RAS, Pushchino, Moscow Region 142290, Russia.
| | - Natalia P Komelina
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia
| | - Maxim O Khrenov
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia
| | | |
Collapse
|
10
|
Guo S, Cosky E, Li F, Guan L, Ji Y, Wei W, Peng C, Geng X, Ding Y. An inhibitory and beneficial effect of chlorpromazine and promethazine (C + P) on hyperglycolysis through HIF-1α regulation in ischemic stroke. Brain Res 2021; 1763:147463. [PMID: 33811844 DOI: 10.1016/j.brainres.2021.147463] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND After ischemic stroke, the increased catabolism of glucose (hyperglycolysis) results in the production of reactive oxygen species (ROS) via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). A depressive or hibernation-like effect of C + P on brain activity was reported to induce neuroprotection. The current study assesses the effect of C + P on hyperglycolysis and NOX activation. METHODS Adult male Sprague-Dawley rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) followed by 6 or 24 h of reperfusion. At the onset of reperfusion, rats received C + P with or without temperature control, or phloretin [glucose transporter (GLUT)-1 inhibitor], or cytochalasin B (GLUT-3 inhibitor). We detected brain ROS, apoptotic cell death, and ATP levels along with HIF-1α expression. Cerebral hyperglycolysis was measured by glucose, protein expression of GLUT-1/3, and phosphofructokinase-1 (PFK-1), as well as lactate and lactate dehydrogenase (LDH) at 6 and 24 h of reperfusion. The enzymatic activity of NOX and protein expression of its subunits (gp91phox) were detected. Neural SHSY5Y cells were placed under 2 h of oxygen-glucose deprivation (OGD) followed by reoxygenation for 6 and 24 h with C + P treatment. Cell viability and protein levels of HIF-1α, GLUT-1/3, PFK-1, LDH, and gp91phox were measured. A HIF-1α overexpression vector was transfected into the cells, and then protein levels of HIF-1α, GLUT-1/3, PFK-1, and LDH were quantitated. In sham-operated rats and control cells, the protein levels of HIF-1α, GLUT-1/3, PFK-1, LDH, and gp91phox were measured at 6 and 24 h after C + P administration. RESULTS C + P reduced the protein elevations after stroke in HIF-1α, glycolytic enzymes, as well as in ROS, cell death, glucose and lactate, but raised ATP levels in the brain. In ischemic rats exposed to GLUT-1/3 inhibitors, ROS, cell death, glucose, and lactate were all decreased, as well as GLUT-1, GLUT-3, LDH, and PFK-1 protein levels. C + P decreased ischemia-induced NOX activation by reducing the enzymatic activity and protein expression of the NOX subunit gp91phox, as was observed in the presence of GLUT-1/3 inhibitors. These markers were significantly decreased following C + P administration with the induced hypothermia, while C + P administration with temperature control at 37 °C induced lesser protection after ischemia stroke. In the OGD/reoxygenation model, C + P treatment increased cell viability and diminished protein levels of HIF-1α, GLUT-1, GLUT-3, PFK-1, LDH, and gp91phox. However, in OGD with HIF-1α overexpression, C + P was unable to effectively reduce the upregulated GLUT-1, GLUT-3, and LDH. In normal conditions, C + P reduced HIF-1α and the levels of key glycolytic enzymes depending on its pharmacological effect. CONCLUSION C + P, partially depending on hypothermia, attenuates hyperglycolysis and NOX activation through HIF-1α regulation.
Collapse
Affiliation(s)
- Sichao Guo
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Eric Cosky
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Fengwu Li
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China
| | - Longfei Guan
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Yu Ji
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Wenjing Wei
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Changya Peng
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA.
| |
Collapse
|
11
|
Shimaoka H, Shiina T, Suzuki H, Horii Y, Horii K, Shimizu Y. Successful induction of deep hypothermia by isoflurane anesthesia and cooling in a non-hibernator, the rat. J Physiol Sci 2021; 71:10. [PMID: 33784982 PMCID: PMC10717611 DOI: 10.1186/s12576-021-00794-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 03/03/2021] [Indexed: 11/10/2022]
Abstract
The aim of the present study was to establish a novel method for inducing deep hypothermia in rats. Cooling rats anesthetized with isoflurane caused a time-dependent decrease in rectal temperature, but cardiac arrest occurred before their body temperature reached 20 °C when isoflurane inhalation was continued during the cooling process. Stopping inhalation of isoflurane when the rectal temperature reached 22.5 °C successfully induced deep hypothermia, although stopping the inhalation at 27.5 °C resulted in spontaneous recovery of rectal temperature. The hypothermic condition was able to be maintained for up to 6 h. A large number of c-Fos-positive cells were detected in the hypothalamus during hypothermia. Both the maintenance of and recovery from hypothermia caused organ injury, but the damage was transient and recovered within 1 week. These findings indicate that the established procedure is appropriate for inducing deep hypothermia without accompanying serious organ injury in rats.
Collapse
Affiliation(s)
- Hiroki Shimaoka
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Takahiko Shiina
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, 1-1 Yanagido, Gifu, 501-1193, Japan.
| | - Hayato Suzuki
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yuuki Horii
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kazuhiro Horii
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yasutake Shimizu
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, 1-1 Yanagido, Gifu, 501-1193, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
12
|
Lv S, Zhao W, Rajah GB, Dandu C, Cai L, Cheng Z, Duan H, Dai Q, Geng X, Ding Y. Rapid Intervention of Chlorpromazine and Promethazine for Hibernation-Like Effect in Stroke: Rationale, Design, and Protocol for a Prospective Randomized Controlled Trial. Front Neurol 2021; 12:621476. [PMID: 33815250 PMCID: PMC8010657 DOI: 10.3389/fneur.2021.621476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Following an acute ischemic stroke (AIS), rapidly initiated reperfusion therapies [i. e., intravenous thrombolysis (IVT) and endovascular treatment (EVT)] demonstrate robust clinical efficacy. However, only a subset of these patients can benefit from these therapies due to their short treatment windows and potential complications. In addition, many patients despite successful reperfusion still have unfavorable outcomes. Thus, neuroprotection strategies are urgently needed for AIS patients. Chlorpromazine and promethazine (C+P) have been employed in clinical practice for antipsychotic and sedative purposes. A clinical study has also shown a neuroprotective effect of C+P on patients with cerebral hemorrhage and subarachnoid hemorrhage. The safety, feasibility, and preliminary efficacy of intravenous administration of C+P in AIS patients within 24 h of onset will be elucidated. Methods: A prospective randomized controlled trial is proposed with AIS patients. Participants will be randomly allocated to an intervention group and a control group with a 1:1 ratio (n = 30) and will be treated with standard therapies according to the current stroke guidelines. Participants allocated to the intervention group will receive intravenous administration of C+P (chlorpromazine 50 mg and promethazine 50 mg) within 24 h of symptom onset. The primary outcome is safety (mainly hypotension), while the secondary outcomes include changes in functional outcome and infarction volume. Discussions: This study on Rapid Intervention of Chlorpromazine and Promethazine for Hibernation-like Effect in Stroke (RICHES) will be the first prospective randomized controlled trial to ascertain the safety, feasibility, and preliminary efficacy of intravenous C+P as a neuroprotection strategy in AIS patients. These results will provide parameters for future studies, provide insights into treatment effects, and neuroprotection with phenothiazine in AIS. Clinical Trial Registration:www.chictr.org.cn, identifier: ChiCTR2000038727.
Collapse
Affiliation(s)
- Shuyu Lv
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Wenbo Zhao
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gary B Rajah
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States.,Department of Neurosurgery, Munson Medical Center, Traverse City, MI, United States
| | - Chaitu Dandu
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Lipeng Cai
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhe Cheng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Honglian Duan
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Qingqing Dai
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States.,Department of Research and Development Center, John D. Dingell VA Medical Center, Detroit, MI, United States
| |
Collapse
|
13
|
Matsuo T, Isosaka T, Tang L, Soga T, Kobayakawa R, Kobayakawa K. Artificial hibernation/life-protective state induced by thiazoline-related innate fear odors. Commun Biol 2021; 4:101. [PMID: 33483561 PMCID: PMC7822961 DOI: 10.1038/s42003-020-01629-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022] Open
Abstract
Innate fear intimately connects to the life preservation in crises, although this relationships is not fully understood. Here, we report that presentation of a supernormal innate fear inducer 2-methyl-2-thiazoline (2MT), but not learned fear stimuli, induced robust systemic hypothermia/hypometabolism and suppressed aerobic metabolism via phosphorylation of pyruvate dehydrogenase, thereby enabling long-term survival in a lethal hypoxic environment. These responses exerted potent therapeutic effects in cutaneous and cerebral ischemia/reperfusion injury models. In contrast to hibernation, 2MT stimulation accelerated glucose uptake in the brain and suppressed oxygen saturation in the blood. Whole-brain mapping and chemogenetic activation revealed that the sensory representation of 2MT orchestrates physiological responses via brain stem Sp5/NST to midbrain PBN pathway. 2MT, as a supernormal stimulus of innate fear, induced exaggerated, latent life-protective effects in mice. If this system is preserved in humans, it may be utilized to give rise to a new field: "sensory medicine."
Collapse
Affiliation(s)
- Tomohiko Matsuo
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoko Isosaka
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan
| | - Lijun Tang
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Reiko Kobayakawa
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan.
| | - Ko Kobayakawa
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan.
| |
Collapse
|
14
|
Shu L, Shen X, Zhao Y, Zhao R, He X, Yin J, Su J, Li Q, Liu J. Mechanisms of transformation of nicotinamide mononucleotides to cerebral infarction hemorrhage based on MCAO model. Saudi J Biol Sci 2020; 27:899-904. [PMID: 32127769 PMCID: PMC7042676 DOI: 10.1016/j.sjbs.2019.12.023] [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] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/16/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The study aims at discussing the effect of nicotinamide mononucleotides on protecting hemorrhagic transformation of cerebral infarction in the middle cerebral artery occlusion (MCAO) model. METHOD Male mice aged 4-5 weeks and weighing about 22-35 g in Shanghai Ninth People's Hospital are divided into three groups: sham group, collagenase intracerebral hemorrhage model (cICH + Vehicle) group and collagenase nicotinamide mononucleotide (cICH + NMN) group. Then, the intervention therapy research is carried out. After 24 h, the neurological function, brain edema, hematoma volume, body weight, hemorrhage volume, RNA expression level, apoptosis, inflammatory factors and reactive oxygen species (ROS) content in surrounding tissues of mice are analyzed comprehensively. RESULTS Compared with the other two groups, nicotinamide mononucleotides in MCAO model have significant effects on improving neurological function, brain edema, inflammatory factors, body weight and cell apoptosis in mice, but have no significant effect on hemorrhage volume and hematoma volume in mice. CONCLUSION Nicotinamide mononucleotides can significantly improve the collagenase-induced intracerebral hemorrhage (ICH) model in mice under MCAO model, and they can protect the brain tissue of mice from RNA level to tissue cell level or mouse body weight and volume level.
Collapse
Affiliation(s)
- Liang Shu
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaolei Shen
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yaxue Zhao
- Department of Neurology, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Rong Zhao
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xinwei He
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jiawen Yin
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jingjing Su
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qiang Li
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jianren Liu
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
15
|
Avogaro A, Fadini GP, Del Prato S. Reinterpreting Cardiorenal Protection of Renal Sodium-Glucose Cotransporter 2 Inhibitors via Cellular Life History Programming. Diabetes Care 2020; 43:501-507. [PMID: 31843950 DOI: 10.2337/dc19-1410] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/03/2019] [Indexed: 02/03/2023]
Abstract
Cardiovascular outcome trials have provided evidence that sodium-glucose cotransporter 2 inhibitor (SGLT2i) treatment is associated with remarkably favorable cardiovascular outcomes. Here, we offer a novel hypothesis that may encompass many of these hypothetical mechanisms, i.e., the ability of SGLT2i to modify the trajectory of cell response to a toxic environment through modifications of cellular life history programs, either the defense program or the dormancy program. The choice between these programs is mainly determined by the environment. Hyperglycemia can be considered a toxic determinant able to interfere with the basic programs of cell evolution. While the defense program is characterized by activation of the immune response and anabolic metabolism, the dormancy program is an energy-preserving state with high resistance to environmental stressors, and it has strong analogy with animal hibernation where fuel is stored, metabolic rate is suppressed, and insulin secretion is reduced. The metabolic changes that follow treatment with SGLT2i are reminiscent of the metabolic picture characteristic of the dormancy program. Therefore, we hypothesize that the beneficial cardioprotective effects of SGLT2i may be related to their ability to switch cell life programming from a defense to a dormancy state, thus lending additional benefit.
Collapse
Affiliation(s)
- Angelo Avogaro
- Section of Diabetes and Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Section of Diabetes and Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy
| | - Stefano Del Prato
- Section of Diabetes and Metabolic Diseases, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
16
|
Tong Y, Elkin KB, Peng C, Shen J, Li F, Guan L, Ji Y, Wei W, Geng X, Ding Y. Reduced Apoptotic Injury by Phenothiazine in Ischemic Stroke through the NOX-Akt/PKC Pathway. Brain Sci 2019; 9:brainsci9120378. [PMID: 31847503 PMCID: PMC6955743 DOI: 10.3390/brainsci9120378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023] Open
Abstract
Phenothiazine treatment has been shown to reduce post-stroke ischemic injury, though the underlying mechanism remains unclear. This study sought to confirm the neuroprotective effects of phenothiazines and to explore the role of the NOX (nicotinamide adenine dinucleotide phosphate oxidase)/Akt/PKC (protein kinase C) pathway in cerebral apoptosis. Sprague-Dawley rats underwent middle cerebral artery occlusion (MCAO) for 2 h and were randomly divided into 3 different cohorts: (1) saline, (2) 8 mg/kg chlorpromazine and promethazine (C+P), and (3) 8 mg/kg C+P as well as apocynin (NOX inhibitor). Brain infarct volumes were examined, and cell death/NOX activity was determined by assays. Western blotting was used to assess protein expression of kinase C-δ (PKC-δ), phosphorylated Akt (p-Akt), Bax, Bcl-XL, and uncleaved/cleaved caspase-3. Both C+P and C+P/NOX inhibitor administration yielded a significant reduction in infarct volumes and cell death, while the C+P/NOX inhibitor did not confer further reduction. In both treatment groups, anti-apoptotic Bcl-XL protein expression generally increased, while pro-apoptotic Bax and caspase-3 proteins generally decreased. PKC protein expression was decreased in both treatment groups, demonstrating a further decrease by C+P/NOX inhibitor at 6 and 24 h of reperfusion. The present study confirms C+P-mediated neuroprotection and suggests that the NOX/Akt/PKC pathway is a potential target for efficacious therapy following ischemic stroke.
Collapse
Affiliation(s)
- Yanna Tong
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
- Department of Neurology, Luhe Clinical Institute, Capital Medical University, Beijing 101100, China
| | - Kenneth B. Elkin
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
| | - Changya Peng
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
| | - Jiamei Shen
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
| | - Fengwu Li
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
| | - Longfei Guan
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
| | - Yu Ji
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
- Department of General Surgery, Luhe Clinical Institute, Capital Medical University, Beijing 101100, China
| | - Wenjing Wei
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
- China-America Institute of Neuroscience, Xuanwu Clinical Institute, Capital Medical University, Beijing 100053, China
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
- Department of Neurology, Luhe Clinical Institute, Capital Medical University, Beijing 101100, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Correspondence: ; Tel.: +86-183-1105-5270
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
| |
Collapse
|
17
|
Guan L, Guo S, Yip J, Elkin KB, Li F, Peng C, Geng X, Ding Y. Artificial Hibernation by Phenothiazines: A Potential Neuroprotective Therapy Against Cerebral Inflammation in Stroke. Curr Neurovasc Res 2019; 16:232-240. [DOI: 10.2174/1567202616666190624122727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022]
Abstract
Background:
The inflammatory response to acute cerebral ischemia is a major factor in
stroke pathobiology and patient outcome. In the clinical setting, no effective pharmacologic treatments
are currently available. Phenothiazine drugs, such as chlorpromazine and promethazine,
(C+P) have been widely studied because of their ability to induce neuroprotection through artificial
hibernation after stroke. The present study determined their effect on the inflammatory response.
Methods:
Sprague-Dawley rats were divided into 4 groups: (1) sham, (2) stroke, (3) stroke treated
by C+P without temperature control and (4) stroke treated by C+P with temperature control (n=8
per group). To assess the neuroprotective effect of C+P, brain damage was measured using infarct
volume and neurological deficits. The expression of inflammatory response molecules tumor necrosis
factor-α (TNF-α), interleukin-1β (IL-1β), intercellular adhesion molecule 1 (ICAM-1), vascular
cell adhesion molecule 1 (VCAM-1), and nuclear factor kappa light chain enhancer of activated
B cells (NF-κB) was determined by real-time PCR and Western blotting
Results:
TNF-α, IL-1β, ICAM-1, VCAM-1, and NF-κB mRNA and protein expressions were upregulated,
and brain damage and neurological deficits were increased after stroke. These markers
of cerebral injury were significantly reduced following C+P administration under drug-induced
hypothermia, while C+P administration under normal body temperature reduced them by a lesser
degree.
Conclusion:
This study showed an inhibitory effect of C+P on brain inflammation, which may be
partially dependent on drug-induced hibernation, as well as other mechanisms of action by these
drugs. These findings further suggest the great potential of C+P in the clinical treatment of ischemic
stroke.
Collapse
Affiliation(s)
- Longfei Guan
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, MI, United States
| | - Sichao Guo
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, MI, United States
| | - James Yip
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, MI, United States
| | - Kenneth B. Elkin
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, MI, United States
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Changya Peng
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, MI, United States
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, MI, United States
| |
Collapse
|
18
|
Gonzalez-Riano C, León-Espinosa G, Regalado-Reyes M, García A, DeFelipe J, Barbas C. Metabolomic Study of Hibernating Syrian Hamster Brains: In Search of Neuroprotective Agents. J Proteome Res 2019; 18:1175-1190. [DOI: 10.1021/acs.jproteome.8b00816] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo, 28223 Pozuelo de Alarcón, Madrid, Spain
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Mamen Regalado-Reyes
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo, 28223 Pozuelo de Alarcón, Madrid, Spain
| | | | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo, 28223 Pozuelo de Alarcón, Madrid, Spain
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Calle de Valderrebollo, 5, 28031 Madrid, Spain
| | | |
Collapse
|
19
|
Li F, Geng X, Yip J, Ding Y. Therapeutic Target and Cell-signal Communication of Chlorpromazine and Promethazine in Attenuating Blood-Brain Barrier Disruption after Ischemic Stroke. Cell Transplant 2018; 28:145-156. [PMID: 30569751 PMCID: PMC6362522 DOI: 10.1177/0963689718819443] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke destroys blood–brain barrier (BBB) integrity. There are currently no effective treatments available in the clinical setting. Post-ischemia treatment with phenothiazine drugs [combined chlorpromazine and promethazine (C+P)] has been shown to be neuroprotective in stroke. The present study determined the effect of C+P in BBB integrity. Sprague-Dawley rats were divided into the following groups (n=8 each): (1) stroke, (2) stroke treated by C+P with temperature control, and (3) stroke treated by C+P without temperature control. Infarct volume and neurological deficits were measured to assess the neuroprotective effect of C+P. BBB permeability was determined by brain edema and Evans blue leakage. Expression of BBB integral molecules, including proteins of aquaporin-4 and -9 (AQP-4, AQP-9), matrix metalloproteinase-2 and -9 (MMP-2, MMP-9), zonula occludens-1 (ZO-1), claudin-1/5, occludin, and laminin were determined by Western blot. Stroke caused brain infarction and neurological deficits, as well as BBB damage, which were all attenuated by C+P through drug-induced hypothermia. When the reduced temperature was controlled to physiological levels, C+P still conferred neuroprotection, suggesting a therapeutic effect independent of hypothermia. Furthermore, C+P significantly attenuated the increase in AQP-4, AQP-9, MMP-2, and MMP-9 levels after stroke, and reversed the decrease in tight junction protein (ZO-1, claudin-1/5, occludin) and basal laminar protein (laminin) levels. This study clearly indicates a beneficial effect of C+P on BBB integrity after stroke, which may be independent of drug-induced hypothermia. These findings further prove the clinical target and cell-signal communication of C+P treatment, which may direct us closer toward the development of an efficacious neuroprotective therapy.
Collapse
Affiliation(s)
- Fengwu Li
- 1 China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- 1 China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.,2 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.,3 Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - James Yip
- 2 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuchuan Ding
- 1 China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.,2 Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
20
|
Ren C, Li S, Rajah G, Shao G, Lu G, Han R, Huang Q, Li H, Ding Y, Jin K, Ji X. Hypoxia, hibernation and Neuroprotection: An Experimental Study in Mice. Aging Dis 2018; 9:761-768. [PMID: 30090664 PMCID: PMC6065299 DOI: 10.14336/ad.2018.0702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/02/2018] [Indexed: 11/01/2022] Open
Abstract
Hibernation is a unique physiological state that evolved to survive periods of food shortages. It is characterized by profound decreases in metabolic rate, body temperature and physiological functions. Studies have shown that animals in hibernation can resist neurological damage. Here, we aimed to study whether hypoxia can induce a hibernation-like state in a traditionally non-hibernating animal and whether it is neuroprotective. All procedures were conducted according to international guidelines on laboratory animal safety. Mice C57BL/6 (19-21g) were placed into a 125 mL jar with fresh air and the jar was sealed with a rubber plug. For each run, the tolerance limit was judged by the animals' appearance for "air hunger". The animal was removed from the jar as soon as its first gasping breath appeared and was moved to another fresh-air-containing jar of similar volume. This procedure was performed in four runs. The hypoxia exposure significantly decreased oxygen (O2) consumption, carbon dioxide (CO2) production, respiratory rate and heart rate. Meanwhile, rectal temperature reached a minimum of 12.7±2.56°C, which is lower than a wide range of ambient temperatures. The mimicked hibernation decreased the infarct size in a focal cerebral ischemia mouse model. Our findings suggest the possibility of inducing suspended animation-like hibernation states for medical applications post injury.
Collapse
Affiliation(s)
- Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Gary Rajah
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Guo Shao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Guowei Lu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Rongrong Han
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Qingjian Huang
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Yuchuan Ding
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kunlin Jin
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| |
Collapse
|
21
|
Zhang J, Liu K, Elmadhoun O, Ji X, Duan Y, Shi J, He X, Liu X, Wu D, Che R, Geng X, Ding Y. Synergistically Induced Hypothermia and Enhanced Neuroprotection by Pharmacological and Physical Approaches in Stroke. Aging Dis 2018; 9:578-589. [PMID: 30090648 PMCID: PMC6065296 DOI: 10.14336/ad.2017.0817] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022] Open
Abstract
Hypothermia is considered as a promising neuroprotective treatment for ischemic stroke but with many limitations. To expand its clinical relevance, this study evaluated the combination of physical (ice pad) and pharmacological [transient receptor potential vanilloid channel 1 (TRPV1) receptor agonist, dihydrocapsaicin (DHC)] approaches for faster cooling and stronger neuroprotection. A total of 144 male Sprague Dawley rats were randomized to 7 groups: sham (n=16), stroke only (n=24), stroke with physical hypothermia at 31ºC for 3 h after the onset of reperfusion (n=24), high-dose DHC (H-DHC)(1.5 mg/kg, n=24), low-dose DHC (L-DHC)(0.5 mg/kg, n=32) with (n=8) or without (n=24) external body temperature control at ~38 ºC (L-DHC, 38 ºC), and combination therapy (L-DHC+ ice pad, n=24). Rats were subjected to middle cerebral artery occlusion (MCAO) for 2 h. Infarct volume, neurological deficits and apoptotic cell death were determined at 24 h after reperfusion. Expression of pro- and anti-apoptotic proteins was evaluated by Western blot. ATP and reactive oxygen species (ROS) were detected by biochemical assays at 6 and 24 h after reperfusion. Combination therapy of L-DHC and ice pad significantly improved every measured outcome compared to monotherapies. Combination therapy achieved hypothermia faster by 28.6% than ice pad, 350% than L-DHC and 200% than H-DHC alone. Combination therapy reduced (p<0.05) neurological deficits by 63% vs. 26% with L-DHC. No effect was observed when using ice pad or H-DHC alone. L-DHC and ice pad combination improved brain oxidative metabolism by reducing (p<0.05) ROS at 6 and 24 h after reperfusion and increasing ATP levels by 42.9% compared to 25% elevation with L-DHC alone. Finally, combination therapy decreased apoptotic cell death by 48.5% vs. 24.9% with L-DHC, associated with increased anti-apoptotic protein and reduced pro-apoptotic protein levels (p<0.001). Our study has demonstrated that combining physical and pharmacological hypothermia is a promising therapeutic approach in ischemic stroke, and warrants further translational investigations.
Collapse
Affiliation(s)
- Jun Zhang
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kaiyin Liu
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Omar Elmadhoun
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xunming Ji
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yunxia Duan
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jingfei Shi
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoduo He
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiangrong Liu
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ruiwen Che
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Jiang X, Gu T, Liu Y, Gao S, Shi E, Zhang G. Chipmunk Brain Is Resistant to Injury from Deep Hypothermic Circulatory Arrest During Cardiopulmonary Bypass. Ther Hypothermia Temp Manag 2018; 9:118-127. [PMID: 30036167 DOI: 10.1089/ther.2018.0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chipmunk as a food-storing hibernator naturally undergoes hibernation that is linked to great changes in systemic physiology and could protect the central nervous system during drastically reduced cerebral blood flow and low temperature in hibernation. Deep hypothermic circulatory arrest (DHCA) is associated with neurological dysfunction. We aim to test whether the euthermic chipmunk is resistant to injury from DHCA. Sprague-Dawley (SD) rats were used in a positive control. Ten euthermic chipmunks and 10 rats were subjected to 60-minute DHCA. Sham rats and chipmunks received cannulations. The blood samples after surgery were extracted to measure the tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) level. The levels of opioid receptor delta 1 (OPRD1), mature brain-derived neurotrophic factor (m-BDNF), precursor of BDNF (pro-BDNF), TrkB, GRB2, Erk, p-Erk, P38, Bcl-2, P75NTR, TRAF6, JNK, P53, Bax, and Caspase3 of the hippocampus were analyzed at 24 hours after surgery. The brain of chipmunks and rats were fixed for histopathological assessment. In the DHCA rat group, the levels of TNF-α and IL-6 were greater (p < 0.05) compared with DHCA chipmunks. In the DHCA chipmunk group, the levels of OPRD1, mature BDNF/pro-BDNF, TrkB-FL/TrkB-T1, Bcl-2, and p-Erk/Erk of hippocampus were higher than DHCA rats. The levels of GRB2, P75NTR, TRAF6, P53, Bax, and Caspase3 in DHCA chipmunks were lower than DHCA rats. The histopathological assessment showed that the injury in DHCA rat group was more severe than the DHCA chipmunk group. Euthermic chipmunks were greatly tolerant to global cerebral injury during DHCA. Different isoforms of BDNF might be involved in the resistant strategy.
Collapse
Affiliation(s)
- Xuan Jiang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yu Liu
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shilun Gao
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Enyi Shi
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Guangwei Zhang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
23
|
Wang H, He Z, Zhang Y, Zhang J. 1 H NMR metabolic signature of cerebrospinal fluid following repetitive lower-limb remote ischemia preconditioning. Neurochem Int 2018; 116:95-103. [DOI: 10.1016/j.neuint.2018.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/03/2018] [Accepted: 02/19/2018] [Indexed: 12/14/2022]
|
24
|
Ou J, Ball JM, Luan Y, Zhao T, Miyagishima KJ, Xu Y, Zhou H, Chen J, Merriman DK, Xie Z, Mallon BS, Li W. iPSCs from a Hibernator Provide a Platform for Studying Cold Adaptation and Its Potential Medical Applications. Cell 2018; 173:851-863.e16. [PMID: 29576452 DOI: 10.1016/j.cell.2018.03.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/12/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022]
Abstract
Hibernating mammals survive hypothermia (<10°C) without injury, a remarkable feat of cellular preservation that bears significance for potential medical applications. However, mechanisms imparting cold resistance, such as cytoskeleton stability, remain elusive. Using the first iPSC line from a hibernating mammal (13-lined ground squirrel), we uncovered cellular pathways critical for cold tolerance. Comparison between human and ground squirrel iPSC-derived neurons revealed differential mitochondrial and protein quality control responses to cold. In human iPSC-neurons, cold triggered mitochondrial stress, resulting in reactive oxygen species overproduction and lysosomal membrane permeabilization, contributing to microtubule destruction. Manipulations of these pathways endowed microtubule cold stability upon human iPSC-neurons and rat (a non-hibernator) retina, preserving its light responsiveness after prolonged cold exposure. Furthermore, these treatments significantly improved microtubule integrity in cold-stored kidneys, demonstrating the potential for prolonging shelf-life of organ transplants. Thus, ground squirrel iPSCs offer a unique platform for bringing cold-adaptive strategies from hibernators to humans in clinical applications. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Jingxing Ou
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John M Ball
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yizhao Luan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Tantai Zhao
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Ophthalmology, The Second Xiang-Ya Hospital, Central South University, Changsha 410011, China
| | - Kiyoharu J Miyagishima
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yufeng Xu
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou 310009, China
| | - Huizhi Zhou
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinguo Chen
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dana K Merriman
- Department of Biology, University of Wisconsin, Oshkosh, WI 54901, USA
| | - Zhi Xie
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Barbara S Mallon
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Rastogi R, Geng X, Li F, Ding Y. NOX Activation by Subunit Interaction and Underlying Mechanisms in Disease. Front Cell Neurosci 2017; 10:301. [PMID: 28119569 PMCID: PMC5222855 DOI: 10.3389/fncel.2016.00301] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase (NOX) is an enzyme complex with the sole function of producing superoxide anion and reactive oxygen species (ROS) at the expense of NADPH. Vital to the immune system as well as cellular signaling, NOX is also involved in the pathologies of a wide variety of disease states. Particularly, it is an integral player in many neurological diseases, including stroke, TBI, and neurodegenerative diseases. Pathologically, NOX produces an excessive amount of ROS that exceed the body’s antioxidant ability to neutralize them, leading to oxidative stress and aberrant signaling. This prevalence makes it an attractive therapeutic target and as such, NOX inhibitors have been studied and developed to counter NOX’s deleterious effects. However, recent studies of NOX have created a better understanding of the NOX complex. Comprised of independent cytosolic subunits, p47-phox, p67-phox, p40-phox and Rac, and membrane subunits, gp91-phox and p22-phox, the NOX complex requires a unique activation process through subunit interaction. Of these subunits, p47-phox plays the most important role in activation, binding and translocating the cytosolic subunits to the membrane and anchoring to p22-phox to organize the complex for NOX activation and function. Moreover, these interactions, particularly that between p47-phox and p22-phox, are dependent on phosphorylation initiated by upstream processes involving protein kinase C (PKC). This review will look at these interactions between subunits and with PKC. It will focus on the interaction involving p47-phox with p22-phox, key in bringing the cytosolic subunits to the membrane. Furthermore, the implication of these interactions as a target for NOX inhibitors such as apocynin will be discussed as a potential avenue for further investigation, in order to develop more specific NOX inhibitors based on the inhibition of NOX assembly and activation.
Collapse
Affiliation(s)
- Radhika Rastogi
- Department of Neurosurgery, Wayne State University School of Medicine Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
26
|
Tarahovsky YS, Fadeeva IS, Komelina NP, Khrenov MO, Zakharova NM. Antipsychotic inductors of brain hypothermia and torpor-like states: perspectives of application. Psychopharmacology (Berl) 2017; 234:173-184. [PMID: 27933367 DOI: 10.1007/s00213-016-4496-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/26/2016] [Indexed: 12/12/2022]
Abstract
Hypothermia and hypometabolism (hypometabothermia) normally observed during natural hibernation and torpor, allow animals to protect their body and brain against the damaging effects of adverse environment. A similar state of hypothermia can be achieved under artificial conditions through physical cooling or pharmacological effects directed at suppression of metabolism and the processes of thermoregulation. In these conditions called torpor-like states, the mammalian ability to recover from stroke, heart attack, and traumatic injuries greatly increases. Therefore, the development of therapeutic methods for different pathologies is a matter of great concern. With the discovery of the antipsychotic drug chlorpromazine in the 1950s of the last century, the first attempts to create a pharmacologically induced state of hibernation for therapeutic purposes were made. That was the beginning of numerous studies in animals and the broad use of therapeutic hypothermia in medicine. Over the last years, many new agents have been discovered which were capable of lowering the body temperature and inhibiting the metabolism. The psychotropic agents occupy a significant place among them, which, in our opinion, is not sufficiently recognized in the contemporary literature. In this review, we summarized the latest achievements related to the ability of modern antipsychotics to target specific receptors in the brain, responsible for the initiation of hypometabothermia.
Collapse
Affiliation(s)
- Yury S Tarahovsky
- Institute of Cell Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290. .,Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290.
| | - Irina S Fadeeva
- Institute of Cell Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290.,Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290
| | - Natalia P Komelina
- Institute of Cell Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290
| | - Maxim O Khrenov
- Institute of Cell Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290
| | - Nadezhda M Zakharova
- Institute of Cell Biophysics RAS, Pushchino, Moscow Region, Russian Federation, 142290
| |
Collapse
|
27
|
Geng X, Li F, Yip J, Peng C, Elmadhoun O, Shen J, Ji X, Ding Y. Neuroprotection by Chlorpromazine and Promethazine in Severe Transient and Permanent Ischemic Stroke. Mol Neurobiol 2016; 54:8140-8150. [DOI: 10.1007/s12035-016-0280-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022]
|
28
|
Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem 2016; 139:1019-1055. [PMID: 27365148 DOI: 10.1111/jnc.13724] [Citation(s) in RCA: 320] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022]
Abstract
The adenosine modulation system mostly operates through inhibitory A1 (A1 R) and facilitatory A2A receptors (A2A R) in the brain. The activity-dependent release of adenosine acts as a brake of excitatory transmission through A1 R, which are enriched in glutamatergic terminals. Adenosine sharpens salience of information encoding in neuronal circuits: high-frequency stimulation triggers ATP release in the 'activated' synapse, which is locally converted by ecto-nucleotidases into adenosine to selectively activate A2A R; A2A R switch off A1 R and CB1 receptors, bolster glutamate release and NMDA receptors to assist increasing synaptic plasticity in the 'activated' synapse; the parallel engagement of the astrocytic syncytium releases adenosine further inhibiting neighboring synapses, thus sharpening the encoded plastic change. Brain insults trigger a large outflow of adenosine and ATP, as a danger signal. A1 R are a hurdle for damage initiation, but they desensitize upon prolonged activation. However, if the insult is near-threshold and/or of short-duration, A1 R trigger preconditioning, which may limit the spread of damage. Brain insults also up-regulate A2A R, probably to bolster adaptive changes, but this heightens brain damage since A2A R blockade affords neuroprotection in models of epilepsy, depression, Alzheimer's, or Parkinson's disease. This initially involves a control of synaptotoxicity by neuronal A2A R, whereas astrocytic and microglia A2A R might control the spread of damage. The A2A R signaling mechanisms are largely unknown since A2A R are pleiotropic, coupling to different G proteins and non-canonical pathways to control the viability of glutamatergic synapses, neuroinflammation, mitochondria function, and cytoskeleton dynamics. Thus, simultaneously bolstering A1 R preconditioning and preventing excessive A2A R function might afford maximal neuroprotection. The main physiological role of the adenosine modulation system is to sharp the salience of information encoding through a combined action of adenosine A2A receptors (A2A R) in the synapse undergoing an alteration of synaptic efficiency with an increased inhibitory action of A1 R in all surrounding synapses. Brain insults trigger an up-regulation of A2A R in an attempt to bolster adaptive plasticity together with adenosine release and A1 R desensitization; this favors synaptotocity (increased A2A R) and decreases the hurdle to undergo degeneration (decreased A1 R). Maximal neuroprotection is expected to result from a combined A2A R blockade and increased A1 R activation. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
29
|
Khoury N, Koronowski KB, Perez-Pinzon MA. Long-term window of ischemic tolerance: An evolutionarily conserved form of metabolic plasticity regulated by epigenetic modifications? ACTA ACUST UNITED AC 2016; 1:6-12. [PMID: 27796011 DOI: 10.29245/2572.942x/2016/2.1021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the absence of effective neuroprotective agents in the clinic, ischemic and pharmacological preconditioning are gaining increased interest in the field of cerebral ischemia. Our lab recently reported that resveratrol preconditioning affords tolerance against a focal cerebral ischemic insult in mice that can last for at least 14 days in vivo making it the longest window of ischemic tolerance discovered to date by a single administration of a pharmacological agent. The mechanism behind this novel extended window of ischemic tolerance remains elusive. In the below commentary we discuss potential mechanisms that could explain this novel extended window of ischemic tolerance in the context of previously identified windows and the known mechanisms behind them. We also draw parallels from the fields of hibernation and hypoxia-tolerance, which are chronic adaptations to severe conditions of hypoxia and ischemia known to be mediated by a form of metabolic depression. We also briefly discuss the importance of epigenetic modifications in maintaining this depressed state of metabolism.
Collapse
Affiliation(s)
- Nathalie Khoury
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Kevin B Koronowski
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|