1
|
Li J, Wang H, Ma P, Li T, Ren J, Zhang J, Zhou M, He Y, Yang T, He W, Mi MT, Liu YW, Dai SS. Osteocalcin-expressing neutrophils from skull bone marrow exert immunosuppressive and neuroprotective effects after TBI. Cell Rep 2024; 43:114670. [PMID: 39213156 DOI: 10.1016/j.celrep.2024.114670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/04/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Neutrophils from skull bone marrow (Nskull) are activated under some brain stresses, but their effects on traumatic brain injury (TBI) are lacking. Here, we find Nskull infiltrates brain tissue quickly and persistently after TBI, which is distinguished by highly and specifically expressed osteocalcin (OCN) from blood-derived neutrophils (Nblood). Reprogramming of glucose metabolism by reducing glycolysis-related enzyme glyceraldehyde 3-phosphate dehydrogenase expression is involved in the antiapoptotic and proliferative abilities of OCN-expressing Nskull. The transcription factor Fos-like 1 governs the specific gene profile of Nskull including C-C motif chemokine receptor-like 2 (CCRL2), arginase 1 (Arg1), and brain-derived neurotrophic factor (BDNF) in addition to OCN. Selective knockout of CCRL2 in Nskull demonstrates that CCRL2 mediates its recruitment, whereas high Arg1 expression is consistent with its immunosuppressive effects on Nblood, and the secretion of BDNF facilitating dendritic growth contributes to its neuroprotection. Thus, our findings provide insight into the roles of Nskull in TBI.
Collapse
Affiliation(s)
- Jiabo Li
- School of Medicine, Chongqing University, Chongqing 400030, China; Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Hao Wang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Pengjiao Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China; Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Health, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Tao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Jiakui Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Jingyu Zhang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Mi Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Yuhang He
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Health, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Teng Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Wenhui He
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Man-Tian Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Health, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China.
| | - Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China.
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
2
|
Wu D, Liao X, Gao J, Gao Y, Li Q, Gao W. Potential pharmaceuticals targeting neuroimmune interactions in treating acute lung injury. Clin Transl Med 2024; 14:e1808. [PMID: 39129233 PMCID: PMC11317502 DOI: 10.1002/ctm2.1808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND AND MAIN BODY Although interactions between the nervous and immune systems have been recognized decades ago, it has become increasingly appreciated that neuroimmune crosstalk is among the driving factors of multiple pulmonary inflammatory diseases including acute lung injury (ALI). Here, we review the current understanding of nerve innervations towards the lung and summarize how the neural regulation of immunity and inflammation participates in the onset and progression of several lung diseases, especially ALI. We then present advancements in the development of potential drugs for ALI targeting neuroimmune interactions, including cholinergic anti-inflammatory pathway, sympathetic-immune pathway, purinergic signalling, neuropeptides and renin-angiotensin system at different stages from preclinical investigation to clinical trials, including the traditional Chinese medicine. CONCLUSION This review highlights the importance of considering the therapeutic strategy of inflammatory diseases within a conceptual framework that integrates classical inflammatory cascade and neuroimmune circuits, so as to deepen the understanding of immune modulation and develop more sophisticated approaches to treat lung diseases represented by ALI. KEY POINTS The lungs present abundant nerve innervations. Neuroimmune interactions exert a modulatory effect in the onset and progression of lung inflammatory diseases, especially acute lung injury. The advancements of potential drugs for ALI targeting neuroimmune crosstalk at different stages from preclinical investigation to clinical trials are elaborated. Point out the direction for the development of neuroimmune pharmacology in the future.
Collapse
Affiliation(s)
- Di Wu
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Ximing Liao
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Jing Gao
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Yixuan Gao
- Department of GynaecologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanP. R. China
| | - Qiang Li
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Wei Gao
- Department of Pulmonary and Critical Care MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghaiP. R. China
| |
Collapse
|
3
|
Li F, Qin N, Yu Y, Dong R, Li X, Gong S, Zeng Z, Huang L, Yang H. TREM-1 inhibition or ondansetron administration ameliorates NLRP3 inflammasome and pyroptosis in traumatic brain injury-induced acute lung injury. Arch Med Sci 2024; 20:984-996. [PMID: 39050170 PMCID: PMC11264077 DOI: 10.5114/aoms/174264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/17/2023] [Indexed: 07/27/2024] Open
Abstract
Introduction Recently, NLR family pyrin domain containing 3 (NLRP3) and pyroptosis have been reported to be involved in traumatic brain injury-induced acute lung injury (TBI-ALI). Studies have shown that triggering receptor expressed on myeloid cells-1 (TREM-1) may be one of the upstream molecules regulating NLRP3/pyroptosis, and 5-hydroxytryptamine type 3-receptor (5-HT3R) antagonists can inhibit NLRP3/pyroptosis. However, the role of TRME-1 in TBI-ALI, the therapeutic effect of 5-HT3R inhibition on TBI-ALI and its mechanism are still unclear. Therefore, this study aimed to evaluate the protective effect of ondansetron, a 5-HT3 inhibitor, on TBI-ALI, and to explore whether the underlying mechanism is related to the regulation of TREM-1. Material and methods A TBI-ALI rat model was constructed via lateral fluid percussion (LFP) brain injury, and either TREM-1 inhibitor (LP17) or ondansetron was administered as needed. Results TBI induced NLRP3 inflammasome, pyroptosis, and TREM-1 activation in rat lung tissues in a time-dependent manner. Inhibition of TREM-1 activity attenuated TBI-ALI; this is evident from reduced pathological scores, wet/dry ratios, and bronchoalveolar lavage fluid protein levels and alleviated NLRP3 inflammasome/pyroptosis. In addition, ondansetron reduced NLRP3 inflammasome/pyroptosis and alleviated TBI-ALI. Moreover, ondansetron reduced TREM-1 activation in macrophages and lung tissue. Conclusions Ondansetron alleviated TBI-ALI. In terms of mechanism, TREM-1 promotes TBI-ALI via the NLRP3-related pyroptosis pathway, and the protective effect of ondansetron on TBI-ALI may be related to the inhibition of TREM-1.
Collapse
Affiliation(s)
- Fen Li
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Na Qin
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Yiqin Yu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Rui Dong
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Xiaojie Li
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Shenhai Gong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, China
| | - Lin Huang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, China
| | - Hong Yang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third Clinical College of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, China
| |
Collapse
|
4
|
Du H, Li C, Gao R, Tan Y, Wang B, Peng Y, Yang N, Ning Y, Li P, Zhao Y, Zhou Y. Inhibition of the interaction between microglial adenosine 2A receptor and NLRP3 inflammasome attenuates neuroinflammation posttraumatic brain injury. CNS Neurosci Ther 2024; 30:e14408. [PMID: 37564004 PMCID: PMC10805470 DOI: 10.1111/cns.14408] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
AIMS Adenosine 2A receptor (A2A R) is widely expressed in the brain and plays important roles in neuroinflammation, and the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a crucial component of the innate immune system while the regulation of A2A R on it in the central nervous system (CNS) has not been clarified. METHODS The effects of microglial A2A R on NLRP3 inflammasome assembly and activation were investigated in wild-type, A2A R- or NLRP3-knockout primary microglia with pharmacological treatment. Microglial A2A R or NLRP3 conditional knockout mice were used to interrogate the effects of this regulation on neuroinflammation posttraumatic brain injury (TBI). RESULTS We found that A2A R directly interacted with NLRP3 and facilitated NLRP3 inflammasome assembly and activation in primary microglia while having no effects on mRNA levels of inflammasome components. Inhibition of the interaction via A2A R agonist or knockout attenuated inflammasome assembly and activation in vitro. In the TBI model, microglial A2A R and NLRP3 were co-expressed at high levels in microglia next to the peri-injured cortex, and abrogating of this interaction by microglial NLRP3 or A2A R conditional knockout attenuated the neurological deficits and neuropathology post-TBI via reducing the NLRP3 inflammasome activation. CONCLUSION Our results demonstrated that inhibition of the interaction between A2A R and NLRP3 in microglia could mitigate the NLRP3 inflammasome assembly and activation and ameliorate the neuroinflammation post-TBI. It provides new insights into the effects of A2A R on neuroinflammation regulation post-TBI and offers a potential target for the treatment of NLRP3 inflammasome-related CNS diseases.
Collapse
Affiliation(s)
- Hao Du
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- The General Hospital of Tibet Military CommandTibetChina
| | - Chang‐Hong Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Ruo‐Bing Gao
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Yan Tan
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Department of Pathophysiology, College of High‐Altitude Military MedicineArmy Medical UniversityChongqingChina
| | - Bo Wang
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Yan Peng
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Nan Yang
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Ya‐Lei Ning
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| | - Ping Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| | - Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| | - Yuan‐Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| |
Collapse
|
5
|
Ruf WP, Palmer A, Dörfer L, Wiesner D, Buck E, Grozdanov V, Kassubek J, Dimou L, Ludolph AC, Huber-Lang M, Danzer KM. Thoracic trauma promotes alpha-Synuclein oligomerization in murine Parkinson's disease. Neurobiol Dis 2022; 174:105877. [PMID: 36162738 DOI: 10.1016/j.nbd.2022.105877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Systemic and neuroinflammatory processes play key roles in neurodegenerative diseases such as Parkinson's disease (PD). Physical trauma which induces considerable systemic inflammatory responses, represents an evident environmental factor in aging. However, little is known about the impact of physical trauma, on the immuno-pathophysiology of PD. Especially blunt chest trauma which is associated with a high morbidity and mortality rate in the elderly population, can induce a strong pulmonary and systemic inflammatory reaction. Hence, we sought out to combine a well-established thoracic trauma mouse model with a well-established PD mouse model to characterize the influence of physical trauma to neurodegenerative processes in PD. METHODS To study the influence of peripheral trauma in a PD mouse model we performed a highly standardized blunt thorax trauma in a well-established PD mouse model and determined the subsequent local and systemic response. RESULTS We could show that blunt chest trauma leads to a systemic inflammatory response which is quantifiable with increased inflammatory markers in bronchoalveolar fluids (BALF) and plasma regardless of the presence of a PD phenotype. A difference of the local inflammatory response in the brain between the PD group and non-PD group could be detected, as well as an increase in the formation of oligomeric pathological alpha-Synuclein (asyn) suggesting an interplay between peripheral thoracic trauma and asyn pathology in PD. CONCLUSION Taken together this study provides evidence that physical trauma is associated with increased asyn oligomerization in a PD mouse model underlining the relevance of PD pathogenesis under traumatic settings.
Collapse
Affiliation(s)
- Wolfgang P Ruf
- Department of Neurology, Ulm University, 89081 Ulm, Germany.
| | - Annette Palmer
- Institute of Experimental Trauma- Immunology, University Hospital of Ulm, Ulm, Germany.
| | - Lena Dörfer
- Institute of Experimental Trauma- Immunology, University Hospital of Ulm, Ulm, Germany
| | - Diana Wiesner
- German Center for Neurodegenerative Diseases (DNZE), 89081 Ulm, Germany.
| | - Eva Buck
- German Center for Neurodegenerative Diseases (DNZE), 89081 Ulm, Germany.
| | | | - Jan Kassubek
- Department of Neurology, Ulm University, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DNZE), 89081 Ulm, Germany.
| | - Leda Dimou
- Molecular and Translational Neuroscience, Ulm University, 89081 Ulm, Germany.
| | - Albert C Ludolph
- Department of Neurology, Ulm University, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DNZE), 89081 Ulm, Germany.
| | - Markus Huber-Lang
- Institute of Experimental Trauma- Immunology, University Hospital of Ulm, Ulm, Germany.
| | - Karin M Danzer
- Department of Neurology, Ulm University, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DNZE), 89081 Ulm, Germany.
| |
Collapse
|
6
|
A 5-HT6R Agonist Alleviates Cognitive Dysfunction after Traumatic Brain Injury in Rats by Increasing BDNF Expression. Behav Brain Res 2022; 433:113997. [DOI: 10.1016/j.bbr.2022.113997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/26/2022] [Accepted: 07/03/2022] [Indexed: 11/22/2022]
|
7
|
Schreiner TG, Popescu BO. Impact of Caffeine on Alzheimer’s Disease Pathogenesis—Protective or Risk Factor? Life (Basel) 2022; 12:life12030330. [PMID: 35330081 PMCID: PMC8952218 DOI: 10.3390/life12030330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), the most common dementia worldwide, remains without an effective treatment to this day despite intensive research conducted during the last decades. In this context, researchers have turned their attention towards the prevention of this pathology, focusing on early detection and better control of the most important risk factors, concomitantly with trying to find potentially protective factors that may delay the onset of AD. From the multitude of factors studied, coffee (especially its main component, caffeine) is a current interesting research topic, taking into consideration the contradictory results of recent years’ studies. On the one hand, much of the evidence from fundamental research suggests the potentially protective trait of caffeine in AD, while other data mainly from human studies lean toward no correlation or even suggesting that caffeine is a veritable risk factor for dementia. Given the methodological heterogeneity of the studies, this review aims to bring new evidence regarding this topic and to try to clearly establish a correlation between the two entities. Thus, in the first part, the authors make a clear distinction between the effects of coffee and the effects of caffeine in AD, presenting a rich basis of clinical trials on both animal models and the human subject. Subsequently, the main pathophysiological mechanisms that would explain the action of caffeine in the etiopathogenesis of AD are reviewed. Finally, the role of computational models is presented, having beneficial impact on both better understanding of the disease mechanism and the development of new therapeutic approaches for AD prevention.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Department of Neurology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
- Correspondence:
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
8
|
Du H, Tan Y, Li CH, Zhao Y, Li P, Ning YL, Gao RB, Wang B, Peng Y, Tan SW, Huang ZZ, Chen X, Yang N, Shan FB, Xiong RP, Zhou YG. High glutamate concentration reverses the inhibitory effect of microglial adenosine 2A receptor on NLRP3 inflammasome assembly and activation. Neurosci Lett 2022; 769:136431. [PMID: 34974110 DOI: 10.1016/j.neulet.2021.136431] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 02/03/2023]
Abstract
NLRP3 inflammasome plays a crucial role in the innate immune system. Our group previously reported that the microglial adenosine 2A receptor (A2AR) regulates canonical neuroinflammation, which is affected by the glutamate concentration. However, the regulatory effect of A2AR on NLRP3 inflammasome and the effects of glutamate concentration remain unknown. Therefore, we aimed to investigate the regulatory effect of microglial A2AR on NLRP3 inflammasome assembly and activation as well as the effects of glutamate concentration on the inflammasome assembly and activation. Experiments were conducted on magnetically sorted primary microglia from P14 mice. The results showed that pharmacological A2AR activation ameliorated NLRP3 activation under no or low glutamate concentrations, but this effect was reversed by high glutamate concentrations. Moreover, the mRNA levels of NLRP3 inflammasome-related genes were not affected by A2AR activation or the glutamate concentration. We further demonstrated that A2AR activation inhibited the interaction between NLRP3 and caspase 1 under no or low glutamate concentrations while promoting their interaction under high glutamate concentrations. The oligomerization of ASC also showed a similar trend. In conclusion, our findings proved that the high glutamate concentration could reverse the inhibition of A2AR on NLRP3 inflammasome activation by modulating its assembly, which provides new insights into the regulatory effect of A2AR on neuroinflammation under different pathological conditions.
Collapse
Affiliation(s)
- Hao Du
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Tan
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Chang-Hong Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China
| | - Ping Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China
| | - Ya-Lei Ning
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China
| | - Ruo-Bing Gao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Bo Wang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Peng
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Si-Wei Tan
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Zhi-Zhong Huang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Xing Chen
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Yang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Fa-Bo Shan
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Ren-Ping Xiong
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, Chongqing, China; Institute of Brain and Intelligence, Army Medical University, Chongqing, China.
| |
Collapse
|
9
|
Tan SW, Zhao Y, Li P, Ning YL, Huang ZZ, Yang N, Liu D, Zhou YG. HMGB1 mediates cognitive impairment caused by the NLRP3 inflammasome in the late stage of traumatic brain injury. J Neuroinflammation 2021; 18:241. [PMID: 34666797 PMCID: PMC8527642 DOI: 10.1186/s12974-021-02274-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/08/2021] [Indexed: 01/01/2023] Open
Abstract
Background Cognitive impairment in the late stage of traumatic brain injury (TBI) is associated with the NOD-, LRR and pyrin domain-containing protein 3 (NLRP3) inflammasome, which plays an important role in neuroinflammation. Although classical inflammatory pathways have been well-documented in the late stage of TBI (4–8 weeks post-injury), the mechanism by which the NLRP3 inflammasome impairs cognition is still unclear. Methods Mice lacking the gene encoding for NLRP3 (NLRP3-knockout mice) and their wild-type littermates were used in a controlled cortical impact model of TBI. Levels of NLRP3 inflammasome and inflammatory factors such as IL-1β and HMGB1 were detected in post-injury hippocampal tissue, as well as long-term potentiation. Behaviors were assessed by T-maze test, novel object recognition, and nesting tests. Glycyrrhizin was used to antagonize HMGB1. Calcium imaging were performed on primary neuronal cultures. Results By using the NLRP3-knockout TBI model, we found that the continuous activation of the NLRP3 inflammasome and high mobility group box 1 (HMGB1) release were closely related to cognitive impairment. We also found that inhibition of HMGB1 improved LTP reduction and cognitive function by increasing the phosphorylation level of the NMDAR1 subunit at serine 896 while reducing NLRP3 inflammasome activation. Conclusion NLRP3 inflammasome damages memory in the late stage of TBI primarily through HMGB1 upregulation and provides an explanation for the long-term progression of cognitive dysfunction. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02274-0.
Collapse
Affiliation(s)
- Si-Wei Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ya-Lei Ning
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhi-Zhong Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Dong Liu
- Medical Center of Trauma and War Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
10
|
Gu Z, Li L, Li Q, Tan H, Zou Z, Chen X, Zhang Z, Zhou Y, Wei D, Liu C, Huang Q, Maegele M, Cai D, Huang M. Polydatin alleviates severe traumatic brain injury induced acute lung injury by inhibiting S100B mediated NETs formation. Int Immunopharmacol 2021; 98:107699. [PMID: 34147911 DOI: 10.1016/j.intimp.2021.107699] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/10/2021] [Accepted: 04/18/2021] [Indexed: 12/28/2022]
Abstract
Severe traumatic brain injury (sTBI)-induced acute lung injury (sTBI-ALI) is regarded as the most common complication of sTBI that is an independent predictor of poor outcomes in patients with sTBI and strongly increases sTBI mortality. Polydatin (PD) has been shown to have a potential therapeutic effect on sTBI-induced neurons injury and sepsis-induced acute lung injury (ALI), therefore, it is reasonable to believe that PD has a protective effect on sTBI-ALI. Here, to clarify the PD protective effect following sTBI-ALI, a rat brain injury model of lateral fluid percussion was established to mimic sTBI. As a result, sTBI induced ALI, and caused an increasing of wet/dry weight ratio and lung vascular permeability, as well as sTBI promoted oxidative stress response in the lung; sTBI caused inflammatory cytokines release, such as IL-6, IL-1β, TNF-α and MCP-1; and sTBI promoted NETs formation, mainly including an increasing expression of MPO, NE and CitH3. Simultaneously, sTBI induced a significant increase in the level of S100B; however, when inhibition of S100B, the expression of MPO, NE and CITH3 were significantly inhibited following sTBI. Inhibition of S100B also promoted lung vascular permeability recovery and alleviated oxidative stress response. Furthermore, PD treatmentreduced the pathological lung damage, promoted lung vascular permeability recovery, alleviated oxidative stress response and inflammatory cytokines release; more importantly, PD inhibited the expression of S100B, and NETs formation in the lung following sTBI. These results indicate that PD alleviates sTBI-ALI by inhibiting S100B mediated NETs formation. Thus, PD may be valuable in sTBI-ALI treatment.
Collapse
Affiliation(s)
- Zhengtao Gu
- Department of Traumatology and Orthopedic Surgery, Shunde Hospital of Southern Medical University, The First People's Hospital of Shunde, Foshan, Guangdong, China; Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Li Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Qin Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Hongping Tan
- Department of Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, Guangdong, China
| | - Zhimin Zou
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Xueyong Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Zichen Zhang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Yijun Zhou
- Department of Orthopaedic , The First people's Hospital of Changde, Guangde Clinical Institute of Xiangya Medical College of South Central University, Changde, Hunan, China
| | - Danian Wei
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Chengyong Liu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Qiaobing Huang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Pathophysiology, Southern Medical University, Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
| | - Marc Maegele
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Treatment Center For Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Traumatology and Orthopedic Surgery, Cologne-Merheim Medical Center (CMMC), University Witten/Herdecke (UW/H), Campus Cologne-Merheim, Cologne, Germany
| | - Daozhang Cai
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China; Department of Orthopedics, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Academy of Orthopedics Guangdong Province, Guangzhou, Guangdong, China.
| | - Mingguang Huang
- Department of Traumatology and Orthopedic Surgery, Shunde Hospital of Southern Medical University, The First People's Hospital of Shunde, Foshan, Guangdong, China.
| |
Collapse
|
11
|
Sun L, Li X, Guan H, Chen S, Fan X, Zhou C, Yang H, Xiao W. A Novel Role of A 2AR in the Maintenance of Intestinal Barrier Function of Enteric Glia from Hypoxia-Induced Injury by Combining with mGluR5. Front Pharmacol 2021; 12:633403. [PMID: 34093180 PMCID: PMC8173626 DOI: 10.3389/fphar.2021.633403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
During acute intestinal ischemia reperfusion (IR) injury, the intestinal epithelial barrier (IEB) function is often disrupted. Enteric glial cells (EGCs) play an important role in maintaining the integrity of IEB functions. However, how EGCs regulate IEB function under IR stimulation is unknown. The present study reveals that the adenosine A2A receptor (A2AR) is important for mediating the barrier-modulating roles of EGCs. A2AR knockout (KO) experiments revealed more serious intestinal injury in A2AR KO mice than in WT mice after IR stimulation. Moreover, A2AR expression was significantly increased in WT mice when challenged by IR. To further investigate the role of A2AR in IEB, we established an in vitro EGC-Caco-2 co-culture system. Hypoxia stimulation was used to mimic the process of in vivo IR. Treating EGCs with the CGS21680 A2AR agonist attenuated hypoxia-induced intestinal epithelium damage through up-regulating ZO-1 and occludin expression in cocultured Caco-2 monolayers. Furthermore, we showed that A2AR and metabotropic glutamate receptor 5 (mGluR5) combine to activate the PKCα-dependent pathway in conditions of hypoxia. This study shows, for the first time, that hypoxia induces A2AR-mGluR5 interaction in EGCs to protect IEB function via the PKCα pathway.
Collapse
Affiliation(s)
- Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiang Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Haidi Guan
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shuaishuai Chen
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xin Fan
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chao Zhou
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Zeng XJ, Li P, Ning YL, Zhao Y, Peng Y, Yang N, Xu YW, Chen JF, Zhou YG. A 2A R inhibition in alleviating spatial recognition memory impairment after TBI is associated with improvement in autophagic flux in RSC. J Cell Mol Med 2020; 24:7000-7014. [PMID: 32394486 PMCID: PMC7299719 DOI: 10.1111/jcmm.15361] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/12/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
Spatial recognition memory impairment is an important complication after traumatic brain injury (TBI). We previously found that spatial recognition memory impairment can be alleviated in adenosine A2A receptor knockout (A2AR KO) mice after TBI, but the mechanism remains unclear. In the current study, we used manganese‐enhanced magnetic resonance imaging and the Y‐maze test to determine whether the electrical activity of neurons in the retrosplenial cortex (RSC) was reduced and spatial recognition memory was impaired in wild‐type (WT) mice after moderate TBI. Furthermore, spatial recognition memory was damaged by optogenetically inhibiting the electrical activity of RSC neurons in WT mice. Additionally, the electrical activity of RSC neurons was significantly increased and spatial recognition memory impairment was reduced in A2AR KO mice after moderate TBI. Specific inhibition of A2AR in the ipsilateral RSC alleviated the impairment in spatial recognition memory in WT mice. In addition, A2AR KO improved autophagic flux in the ipsilateral RSC after injury. In primary cultured neurons, activation of A2AR reduced lysosomal‐associated membrane protein 1 and cathepsin D (CTSD) levels, increased phosphorylated protein kinase A and phosphorylated extracellular signal‐regulated kinase 2 levels, reduced transcription factor EB (TFEB) nuclear localization and impaired autophagic flux. These results suggest that the impairment of spatial recognition memory after TBI may be associated with impaired autophagic flux in the RSC and that A2AR activation may reduce lysosomal biogenesis through the PKA/ERK2/TFEB pathway to impair autophagic flux.
Collapse
Affiliation(s)
- Xu-Jia Zeng
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Li
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ya-Lei Ning
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Zhao
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Peng
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Yang
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ya-Wei Xu
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiang-Fan Chen
- Department of Neurology and Pharmacology, Boston University School of Medicine, Boston, MA, USA
| | - Yuan-Guo Zhou
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
13
|
Reduction in Blood Glutamate Levels Combined With the Genetic Inactivation of A2AR Significantly Alleviate Traumatic Brain Injury-Induced Acute Lung Injury. Shock 2020; 51:502-510. [PMID: 29688987 DOI: 10.1097/shk.0000000000001170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Traumatic brain injury-induced acute lung injury (TBI-ALI) is a serious complication of traumatic brain injury (TBI). Our previous clinical study found that high levels of blood glutamate after TBI were closely related to the occurrence and severity of TBI-ALI, while it remains unknown whether a high concentration of blood glutamate directly causes or aggravates TBI-ALI. We found that inhibition of the adenosine A2A receptor (A2AR) after brain injury alleviated the TBI-ALI; however, it is unknown whether lowering blood glutamate levels in combination with inhibiting the A2AR would lead to better effects. Using mouse models of moderate and severe TBI, we found that intravenous administration of L-glutamate greatly increased the lung water content, lung-body index, level of inflammatory markers in bronchoalveolar lavage fluid and acute lung injury score and significantly decreased the PaO2/FiO2 ratio. Moreover, the incidence of TBI-ALI and the mortality rate were significantly increased, and the combined administration of A2AR activator and exogenous glutamate further exacerbated the above damaging effects. Conversely, lowering the blood glutamate level through peritoneal dialysis or intravenous administration of oxaloacetate notably improved the above parameters, and a further improvement was seen with concurrent A2AR genetic inactivation. These data suggest that A2AR activation aggravates the damaging effect of high blood glutamate concentrations on the lung and that combined treatment targeting both A2AR and blood glutamate may be an effective way to prevent and treat TBI-ALI.
Collapse
|
14
|
Tasker RC. Traumatic Brain Injury and Pediatric Acute Respiratory Distress Syndrome: Moving the Field Forward. Pediatr Crit Care Med 2020; 21:198-199. [PMID: 32032266 DOI: 10.1097/pcc.0000000000002181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Robert C Tasker
- Department of Neurology; and Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School and Boston Children's Hospital, Boston, MA
| |
Collapse
|
15
|
Sasannejad C, Ely EW, Lahiri S. Long-term cognitive impairment after acute respiratory distress syndrome: a review of clinical impact and pathophysiological mechanisms. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:352. [PMID: 31718695 PMCID: PMC6852966 DOI: 10.1186/s13054-019-2626-z] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) survivors experience a high prevalence of cognitive impairment with concomitantly impaired functional status and quality of life, often persisting months after hospital discharge. In this review, we explore the pathophysiological mechanisms underlying cognitive impairment following ARDS, the interrelations between mechanisms and risk factors, and interventions that may mitigate the risk of cognitive impairment. Risk factors for cognitive decline following ARDS include pre-existing cognitive impairment, neurological injury, delirium, mechanical ventilation, prolonged exposure to sedating medications, sepsis, systemic inflammation, and environmental factors in the intensive care unit, which can co-occur synergistically in various combinations. Detection and characterization of pre-existing cognitive impairment imparts challenges in clinical management and longitudinal outcome study enrollment. Patients with brain injury who experience ARDS constitute a distinct population with a particular combination of risk factors and pathophysiological mechanisms: considerations raised by brain injury include neurogenic pulmonary edema, differences in sympathetic activation and cholinergic transmission, effects of positive end-expiratory pressure on cerebral microcirculation and intracranial pressure, and sensitivity to vasopressor use and volume status. The blood-brain barrier represents a physiological interface at which multiple mechanisms of cognitive impairment interact, as acute blood-brain barrier weakening from mechanical ventilation and systemic inflammation can compound existing chronic blood-brain barrier dysfunction from Alzheimer’s-type pathophysiology, rendering the brain vulnerable to both amyloid-beta accumulation and cytokine-mediated hippocampal damage. Although some contributory elements, such as the presenting brain injury or pre-existing cognitive impairment, may be irreversible, interventions such as minimizing mechanical ventilation tidal volume, minimizing duration of exposure to sedating medications, maintaining hemodynamic stability, optimizing fluid balance, and implementing bundles to enhance patient care help dramatically to reduce duration of delirium and may help prevent acquisition of long-term cognitive impairment.
Collapse
Affiliation(s)
- Cina Sasannejad
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - E Wesley Ely
- Critical Illness, Brain Dysfunction, Survivorship (CIBS) Center, Department of Pulmonary and Critical Care Medicine, Veteran's Affairs Tennessee Valley Geriatric Research Education and Clinical Center (GRECC), Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shouri Lahiri
- Division of Neurocritical Care, Department of Neurology, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd, AHSP Building, Suite A6600, A8103, Los Angeles, CA, 90048, USA. .,Division of Neurocritical Care, Department of Neurosurgery, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd, AHSP Building, Suite A6600, A8103, Los Angeles, CA, 90048, USA. .,Division of Neurocritical Care, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd, AHSP Building, Suite A6600, A8103, Los Angeles, CA, 90048, USA.
| |
Collapse
|
16
|
Choi WM, Kim HH, Kim MH, Cinar R, Yi HS, Eun HS, Kim SH, Choi YJ, Lee YS, Kim SY, Seo W, Lee JH, Shim YR, Kim YE, Yang K, Ryu T, Hwang JH, Lee CH, Choi HS, Gao B, Kim W, Kim SK, Kunos G, Jeong WI. Glutamate Signaling in Hepatic Stellate Cells Drives Alcoholic Steatosis. Cell Metab 2019; 30:877-889.e7. [PMID: 31474565 PMCID: PMC6834910 DOI: 10.1016/j.cmet.2019.08.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/15/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022]
Abstract
Activation of hepatocyte cannabinoid receptor-1 (CB1R) by hepatic stellate cell (HSC)-derived 2-arachidonoylglycerol (2-AG) drives de novo lipogenesis in alcoholic liver disease (ALD). How alcohol stimulates 2-AG production in HSCs is unknown. Here, we report that chronic alcohol consumption induced hepatic cysteine deficiency and subsequent glutathione depletion by impaired transsulfuration pathway. A compensatory increase in hepatic cystine-glutamate anti-porter xCT boosted extracellular glutamate levels coupled to cystine uptake both in mice and in patients with ALD. Alcohol also induced the selective expression of metabotropic glutamate receptor-5 (mGluR5) in HSCs where mGluR5 activation stimulated 2-AG production. Consistently, genetic or pharmacologic inhibition of mGluR5 or xCT attenuated alcoholic steatosis in mice via the suppression of 2-AG production and subsequent CB1R-mediated de novo lipogenesis. We conclude that a bidirectional signaling operates at a metabolic synapse between hepatocytes and HSCs through xCT-mediated glutamate-mGluR5 signaling to produce 2-AG, which induces CB1R-mediated alcoholic steatosis.
Collapse
Affiliation(s)
- Won-Mook Choi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hee-Hoon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Myung-Ho Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hyon-Seung Yi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Internal Medicine, Chungnam National University, School of Medicine, Daejeon 35015, Republic of Korea
| | - Hyuk Soo Eun
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Internal Medicine, Chungnam National University, School of Medicine, Daejeon 35015, Republic of Korea
| | - Seok-Hwan Kim
- Department of Surgery, Chungnam National University, College of Medicine, Daejeon 35015, Republic of Korea
| | - Young Jae Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Sun Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Internal Medicine, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - So Yeon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Wonhyo Seo
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun-Hee Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Young-Ri Shim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Ye Eun Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Keungmo Yang
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Tom Ryu
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hueng-Sik Choi
- School of the Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
17
|
PKC Mediates LPS-Induced IL-1β Expression and Participates in the Pro-inflammatory Effect of A 2AR Under High Glutamate Concentrations in Mouse Microglia. Neurochem Res 2019; 44:2755-2764. [PMID: 31650360 DOI: 10.1007/s11064-019-02895-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/23/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023]
Abstract
Pathogens such as bacterial lipopolysaccharide (LPS) play an important role in promoting the production of the inflammatory cytokines interleukin-1 beta (IL-1β) and tumour necrosis factor-α (TNF-α) in response to infection or damage in microglia. However, whether different signalling pathways regulate these two inflammatory factors remains unclear. The protein kinase C (PKC) family is involved in the regulation of inflammation, and our previous research showed that the activation of the PKC pathway played a key role in the LPS-induced transformation of the adenosine A2A receptor (A2AR) from anti-inflammatory activity to pro-inflammatory activity under high glutamate concentrations. Therefore, in the current study, we investigated the role of PKC in the LPS-induced production of these inflammatory cytokines in mouse primary microglia. GF109203X, a specific PKC inhibitor, inhibited the LPS-induced expression of IL-1β messenger ribonucleic acid and intracellular protein in a dose-dependent manner. Moreover, 5 µM GF109203X prevented LPS-induced IL-1β expression but did not significantly affect LPS-induced TNF-α expression. PKC promoted IL-1β expression by regulating the activity of NF-κB but did not significantly impact the activity of ERK1/2. A2AR activation by CGS21680, an A2AR agonist, facilitated LPS-induced IL-1β expression through the PKC pathway at high glutamate concentrations but did not significantly affect LPS-induced TNF-α expression. Taken together, these results suggest a new direction for specific intervention with LPS-induced inflammatory factors in response to specific signalling pathways and provide a mechanism for A2AR targeting, especially after brain injury, to influence inflammation by interfering with A2AR.
Collapse
|
18
|
Samary CS, Ramos AB, Maia LA, Rocha NN, Santos CL, Magalhães RF, Clevelario AL, Pimentel-Coelho PM, Mendez-Otero R, Cruz FF, Capelozzi VL, Ferreira TPT, Koch T, de Abreu MG, Dos Santos CC, Pelosi P, Silva PL, Rocco PRM. Focal ischemic stroke leads to lung injury and reduces alveolar macrophage phagocytic capability in rats. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:249. [PMID: 30290827 PMCID: PMC6173845 DOI: 10.1186/s13054-018-2164-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Ischemic stroke causes brain inflammation, which we postulate may result in lung damage. Several studies have focused on stroke-induced immunosuppression and lung infection; however, the possibility that strokes may trigger lung inflammation has been overlooked. We hypothesized that even focal ischemic stroke might induce acute systemic and pulmonary inflammation, thus altering respiratory parameters, lung tissue integrity, and alveolar macrophage behavior. METHODS Forty-eight Wistar rats were randomly assigned to ischemic stroke (Stroke) or sham surgery (Sham). Lung function, histology, and inflammation in the lung, brain, bronchoalveolar lavage fluid (BALF), and circulating plasma were evaluated at 24 h. In vitro, alveolar macrophages from naïve rats (unstimulated) were exposed to serum or BALF from Sham or Stroke animals to elucidate possible mechanisms underlying alterations in alveolar macrophage phagocytic capability. Alveolar macrophages and epithelial and endothelial cells of Sham and Stroke animals were also isolated for evaluation of mRNA expression of interleukin (IL)-6 and tumor necrosis factor (TNF)-α. RESULTS Twenty-four hours following ischemic stroke, the tidal volume, expiratory time, and mean inspiratory flow were increased. Compared to Sham animals, the respiratory rate and duty cycle during spontaneous breathing were reduced, but this did not affect lung mechanics during mechanical ventilation. Lungs from Stroke animals showed clear evidence of increased diffuse alveolar damage, pulmonary edema, and inflammation markers. This was associated with an increase in ultrastructural damage, as evidenced by injury to type 2 pneumocytes and endothelial cells, cellular infiltration, and enlarged basement membrane thickness. Protein levels of proinflammatory mediators were documented in the lung, brain, and plasma (TNF-α and IL-6) and in BALF (TNF-α). The phagocytic ability of macrophages was significantly reduced. Unstimulated macrophages isolated from naïve rats only upregulated expression of TNF-α and IL-6 following exposure to serum from Stroke rats. Exposure to BALF from Stroke or Sham animals did not change alveolar macrophage behavior, or gene expression of TNF-α and IL-6. IL-6 expression was increased in macrophages and endothelial cells from Stroke animals. CONCLUSIONS In rats, focal ischemic stroke is associated with brain-lung crosstalk, leading to increased pulmonary damage and inflammation, as well as reduced alveolar macrophage phagocytic capability, which seems to be promoted by systemic inflammation.
Collapse
Affiliation(s)
- Cynthia S Samary
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Alane B Ramos
- Laboratory of Cellular and Molecular Neurobiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lígia A Maia
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Nazareth N Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Cíntia L Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Raquel F Magalhães
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Amanda L Clevelario
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Pedro M Pimentel-Coelho
- Laboratory of Cellular and Molecular Neurobiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rosália Mendez-Otero
- Laboratory of Cellular and Molecular Neurobiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Vera L Capelozzi
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Tatiana P T Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Thea Koch
- Pulmonary Engineering Group, Department of Anesthesiology and Intensive Care Therapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marcelo Gama de Abreu
- Pulmonary Engineering Group, Department of Anesthesiology and Intensive Care Therapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Paolo Pelosi
- Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate (DISC), Università degli Studi di Genova, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
19
|
Gil A, van der Pol A, van der Meer P, Bischoff R. LC-MS analysis of key components of the glutathione cycle in tissues and body fluids from mice with myocardial infarction. J Pharm Biomed Anal 2018; 160:289-296. [DOI: 10.1016/j.jpba.2018.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022]
|
20
|
Wang Y, Wang C, Zhang D, Wang H, Bo L, Deng X. Dexmedetomidine Protects Against Traumatic Brain Injury-Induced Acute Lung Injury in Mice. Med Sci Monit 2018; 24:4961-4967. [PMID: 30013022 PMCID: PMC6067036 DOI: 10.12659/msm.908133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background Traumatic brain injury (TBI) leads to acute lung injury (ALI), in which the inflammatory response plays an important role in its pathophysiology. Recent studies suggest that dexmedetomidine (Dex) plays a protective role in acute inflammatory diseases. However, whether Dex has a protective effect on TBI-induced ALI is not clear. The aim of this study was to investigate the effect of Dex on TBI-induced ALI in mice. Material/Methods Mice were randomly divided into 5 groups: 1) sham group; 2) TBI group; 3) TBI+Dex group; 4) TBI+atipamezole (Atip) group; and 5) TBI+Dex+Atip group. Dex (50 μg/kg) was intraperitoneal injected immediately after TBI. The α2 adrenergic antagonist Atip (250 μg/kg) was intraperitoneal injected 15 minutes prior to Dex treatment. Then 24 hours later, the protein concentration in the bronchoalveolar lavage fluid (BALF), lung wet to dry weight ratio, hematoxylin and eosin (H&E) staining of lungs, the level of high-mobility group box protein 1(HMGB1) in serum, and the receptor for advanced glycation end products (RAGE) expression in lung were detected. Results Dex ameliorated the score of lung histological examination, as well as the severity of pulmonary edema and permeability. Moreover, Dex was observed to significantly suppress the expression of HMGBI and RAGE. However, the protective effects of Dex were partially reversed by the administration of Atip. Conclusions Dex may protect against TBI-induced ALI via the HMGB1-RAGE signal pathway, and this protective effect is partly dependent on its α2 adrenoceptor agonist action.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland).,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| | - Changli Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Dan Zhang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland).,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| | - Huihui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland).,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| |
Collapse
|
21
|
Feng LR, Fernández-Martínez JL, Zaal KJ, deAndrés-Galiana EJ, Wolff BS, Saligan LN. mGluR5 mediates post-radiotherapy fatigue development in cancer patients. Transl Psychiatry 2018; 8:110. [PMID: 29849049 PMCID: PMC5976668 DOI: 10.1038/s41398-018-0161-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/26/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer-related fatigue (CRF) is a common burden in cancer patients and little is known about its underlying mechanism. The primary aim of this study was to identify gene signatures predictive of post-radiotherapy fatigue in prostate cancer patients. We employed Fisher Linear Discriminant Analysis (LDA) to identify predictive genes using whole genome microarray data from 36 men with prostate cancer. Ingenuity Pathway Analysis was used to determine functional networks of the predictive genes. Functional validation was performed using a T lymphocyte cell line, Jurkat E6.1. Cells were pretreated with metabotropic glutamate receptor 5 (mGluR5) agonist (DHPG), antagonist (MPEP), or control (PBS) for 20 min before irradiation at 8 Gy in a Mark-1 γ-irradiator. NF-κB activation was assessed using a NF-κB/Jurkat/GFP Transcriptional Reporter Cell Line. LDA achieved 83.3% accuracy in predicting post-radiotherapy fatigue. "Glutamate receptor signaling" was the most significant (p = 0.0002) pathway among the predictive genes. Functional validation using Jurkat cells revealed clustering of mGluR5 receptors as well as increased regulated on activation, normal T cell expressed and secreted (RANTES) production post irradiation in cells pretreated with DHPG, whereas inhibition of mGluR5 activity with MPEP decreased RANTES concentration after irradiation. DHPG pretreatment amplified irradiation-induced NF-κB activation suggesting a role of mGluR5 in modulating T cell activation after irradiation. These results suggest that mGluR5 signaling in T cells may play a key role in the development of chronic inflammation resulting in fatigue and contribute to individual differences in immune responses to radiation. Moreover, modulating mGluR5 provides a novel therapeutic option to treat CRF.
Collapse
Affiliation(s)
- Li Rebekah Feng
- 0000 0001 2297 5165grid.94365.3dNational Institute of Nursing Research, National Institutes of Health, Bethesda, MD USA
| | | | - Kristien J.M. Zaal
- 0000 0001 2297 5165grid.94365.3dLight Imaging Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD USA
| | | | - Brian S. Wolff
- 0000 0001 2297 5165grid.94365.3dNational Institute of Nursing Research/National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Leorey N. Saligan
- 0000 0001 2297 5165grid.94365.3dNational Institute of Nursing Research, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
22
|
Liu YW, Li S, Dai SS. Neutrophils in traumatic brain injury (TBI): friend or foe? J Neuroinflammation 2018; 15:146. [PMID: 29776443 PMCID: PMC5960133 DOI: 10.1186/s12974-018-1173-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/23/2018] [Indexed: 12/26/2022] Open
Abstract
Our knowledge of the pathophysiology about traumatic brain injury (TBI) is still limited. Neutrophils, as the most abundant leukocytes in circulation and the first-line transmigrated immune cells at the sites of injury, are highly involved in the initiation, development, and recovery of TBI. Nonetheless, our understanding about neutrophils in TBI is obsolete, and mounting evidences from recent studies have challenged the conventional views. This review summarizes what is known about the relationships between neutrophils and pathophysiology of TBI. In addition, discussions are made on the complex roles as well as the controversial views of neutrophils in TBI.
Collapse
Affiliation(s)
- Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, People's Republic of China.,Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Song Li
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, People's Republic of China. .,Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
23
|
Bai W, Li P, Ning YL, Peng Y, Xiong RP, Yang N, Chen X, Zhou YG. Adenosine A 2A receptor inhibition restores the normal transport of endothelial glutamate transporters in the brain. Biochem Biophys Res Commun 2018. [PMID: 29526759 DOI: 10.1016/j.bbrc.2018.03.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Excitatory amino acid transporters (EAATs) on cerebral vascular endothelial cells play an important role in maintaining glutamate homeostasis in the brain. The dysfunction of endothelial EAATs is an important reason for the dramatically elevated brain glutamate levels after brain injury, such as traumatic brain injury (TBI). The adenosine A2A receptor (A2AR) plays an important role in regulating the brain glutamate level after brain injury; however, researchers have not clearly determined whether this role was related to its ability to regulate endothelial EAATs. Activation of A2AR in vitro not only decreased the PKA- and glutamate level-dependent strengthening of the interaction between NKA-α1 and the FXYD1 subunit and the subsequent decrease in the activity of Na+/K+-ATPases (NKAs) but also enhanced its interaction with EAATs and ultimately aggravated the reverse transport function of endothelial EAATs under oxygen-glucose deprivation (OGD) conditions. Conversely, inhibition of A2AR restored the normal transport of EAAT. Moreover, A2AR inhibition increased NKA activity and decreased its interaction with EAATs in isolated brain capillaries after TBI, further confirming its role in endothelial EAATs in vivo. Based on our results, A2AR played an important role in regulating endothelial EAAT function, and strategies that restore the normal transport of endothelial EAATs through the inhibition of A2AR might serve as an effective treatment for brain injury.
Collapse
Affiliation(s)
- Wei Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ping Li
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ya-Lei Ning
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Peng
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ren-Ping Xiong
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Nan Yang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xing Chen
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
24
|
Abstract
Trauma can affect any individual at any location and at any time over a lifespan. The disruption of macrobarriers and microbarriers induces instant activation of innate immunity. The subsequent complex response, designed to limit further damage and induce healing, also represents a major driver of complications and fatal outcome after injury. This Review aims to provide basic concepts about the posttraumatic response and is focused on the interactive events of innate immunity at frequent sites of injury: the endothelium at large, and sites within the lungs, inside and outside the brain and at the gut barrier.
Collapse
|
25
|
Impaired autophagic flux is associated with the severity of trauma and the role of A 2AR in brain cells after traumatic brain injury. Cell Death Dis 2018; 9:252. [PMID: 29449536 PMCID: PMC5833790 DOI: 10.1038/s41419-018-0316-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/10/2017] [Accepted: 01/12/2018] [Indexed: 02/06/2023]
Abstract
Recent studies have shown that after traumatic brain injury (TBI), the number of autophagosomes is markedly increased in brain cells surrounding the wound; however, whether autophagy is enhanced or suppressed by TBI remains controversial. In our study, we used a controlled cortical impact system to establish models of mild, moderate and severe TBI. In the mild TBI model, the levels of autophagy-related protein 6 (Beclin1) and autophagy-related protein 12 (ATG12)-autophagy-related protein 5 (ATG5) conjugates were increased, indicating the enhanced initiation of autophagy. Furthermore, the level of the autophagic substrate sequestosome 1 (SQSTM1) was decreased in the ipsilateral cortex. This result, together with the results observed in tandem mRFP-GFP-LC3 adeno-associated virus (AAV)-infected mice, indicates that autophagosome clearance was also increased after mild TBI. Conversely, following moderate and severe TBI, there was no change in the initiation of autophagy, and autophagosome accumulation was observed. Next, we used chloroquine (CQ) to artificially impair autophagic flux in the injured cortex of the mild TBI model and found that the severity of trauma was obviously exacerbated. In addition, autophagic flux and trauma severity were significantly improved in adenosine A2A receptor (A2AR) knockout (KO) mice subjected to moderate TBI. Thus, A2AR may be involved in regulating the impairment of autophagic flux in response to brain injury. Our findings suggest that whether autophagy is increased after TBI is associated with whether autophagic flux is impaired, and the impairment of autophagic flux exacerbates the severity of trauma. Furthermore, A2AR may be a target for alleviating the impairment in autophagic flux after TBI.
Collapse
|
26
|
Bai W, Li W, Ning YL, Li P, Zhao Y, Yang N, Jiang YL, Liang ZP, Jiang DP, Wang Y, Zhang M, Zhou YG. Blood Glutamate Levels Are Closely Related to Acute Lung Injury and Prognosis after Stroke. Front Neurol 2018; 8:755. [PMID: 29403427 PMCID: PMC5785722 DOI: 10.3389/fneur.2017.00755] [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: 10/13/2017] [Accepted: 12/29/2017] [Indexed: 11/13/2022] Open
Abstract
Background Acute lung injury (ALI) is a serious complication of stroke that occurs with a high incidence. Our preclinical results indicated that ALI might be related to blood glutamate levels after brain injury. The purpose of this study was to assess dynamic changes in blood glutamate levels in patients with stroke and to determine the correlation between blood glutamate levels, ALI, and long-term prognosis after stroke. Methods Venous blood samples were collected from controls and patients with stroke at admission and on the third and seventh day after the onset of stroke. Patients were followed for 3 months. The correlations among blood glutamate levels, severities of stroke and ALI, and long-term outcomes were analyzed, and the predictive values of blood glutamate levels and severity scores for ALI were assessed. Results In this study, a total of 384 patients with stroke were enrolled, with a median age of 59 years. Patients showed significantly increased blood glutamate levels within 7 days of stroke onset (p < 0.05), and patients with more severe injuries showed higher blood glutamate levels. Moreover, blood glutamate levels were closely related to the occurrence (adjusted odds ratio, 3.022, p = 0.003) and severity (p < 0.001) of ALI and the long-term prognosis after stroke (p < 0.05), and they were a more accurate predictor of ALI than the more commonly used severity scores (p < 0.01). Conclusion These results indicated that an increased blood glutamate level was closely related to the development of ALI and a poor prognosis after stroke. Clinical Trial Registration http://www.chictr.org.cn, identifier ChiCTR-RPC-15006770.
Collapse
Affiliation(s)
- Wei Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wei Li
- Department of Neurology, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ya-Lei Ning
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ping Li
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Zhao
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Nan Yang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu-Lin Jiang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ze-Ping Liang
- Department of ICU, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Dong-Po Jiang
- Department of ICU, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ying Wang
- Department of Neurology, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Meng Zhang
- Department of Neurology, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
Bai W, Zhou YG. Homeostasis of the Intraparenchymal-Blood Glutamate Concentration Gradient: Maintenance, Imbalance, and Regulation. Front Mol Neurosci 2017; 10:400. [PMID: 29259540 PMCID: PMC5723322 DOI: 10.3389/fnmol.2017.00400] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022] Open
Abstract
It is widely accepted that glutamate is the most important excitatory neurotransmitter in the central nervous system (CNS). However, there is also a large amount of glutamate in the blood. Generally, the concentration gradient of glutamate between intraparenchymal and blood environments is stable. However, this gradient is dramatically disrupted under a variety of pathological conditions, resulting in an amplifying cascade that causes a series of pathological reactions in the CNS and peripheral organs. This eventually seriously worsens a patient’s prognosis. These two “isolated” systems are rarely considered as a whole even though they mutually influence each other. In this review, we summarize what is currently known regarding the maintenance, imbalance and regulatory mechanisms that control the intraparenchymal-blood glutamate concentration gradient, discuss the interrelationships between these systems and further explore their significance in clinical practice.
Collapse
Affiliation(s)
- Wei Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
28
|
Metabotropic glutamate receptor 5 deficiency inhibits neutrophil infiltration after traumatic brain injury in mice. Sci Rep 2017; 7:9998. [PMID: 28855570 PMCID: PMC5577182 DOI: 10.1038/s41598-017-10201-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/07/2017] [Indexed: 11/08/2022] Open
Abstract
Both brain native inflammatory cells and infiltrated peripheral white blood cells (WBCs) are primary participants in the brain inflammatory damage post-TBI. Metabotropic glutamate receptor 5 (mGluR5) has been reported to regulate microglias and astrocytes to affect inflammation after TBI, but its effect on modulating infiltrated peripheral WBCs remains unclear. In a mouse moderate TBI model, we found that mGluR5 knockout (KO) significantly reduced neutrophil infiltration and inflammatory cytokine expression in the brain at 24 hours post TBI, which was accompanied by improved neurological dysfunction. Further investigation indicated that mGluR5 KO reduced the permeability of blood-brain barrier (BBB), the entrance for neutrophils to enter brain, and markedly decreased the mRNA levels of neutrophil-associated chemokines in brain tissue, including CXCL1, CXCL2, CCL2, CCL4 and CCL5. Using brain microvascular endothelial cells (BMECs), neutrophils and a BBB model in vitro, we confirmed the inhibitory effect of mGluR5 deficiency on neutrophil infiltration and demonstrated that blockade of protein kinase C (PKC) signaling was involved in it. These results provide insight into the role of mGluR5 in the regulation of inflammation in the acute phase of TBI, which may provide novel clues for TBI therapy.
Collapse
|
29
|
Bai W, Zhu WL, Ning YL, Li P, Zhao Y, Yang N, Chen X, Jiang YL, Yang WQ, Jiang DP, Chen LY, Zhou YG. Dramatic increases in blood glutamate concentrations are closely related to traumatic brain injury-induced acute lung injury. Sci Rep 2017; 7:5380. [PMID: 28710466 PMCID: PMC5511144 DOI: 10.1038/s41598-017-05574-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/31/2017] [Indexed: 11/27/2022] Open
Abstract
Traumatic brain injury-induced acute lung injury (TBI-ALI) is a serious complication after brain injury for which predictive factors are lacking. In this study, we found significantly elevated blood glutamate concentrations in patients with TBI or multiple peripheral trauma (MPT), and patients with more severe injuries showed higher blood glutamate concentrations and longer durations of elevated levels. Although the increase in amplitude was similar between the two groups, the duration was longer in the patients with TBI. There were no significant differences in blood glutamate concentrations in the patients with MPT with regard to ALI status, but the blood glutamate levels were significantly higher in the patients with TBI-ALI than in those without ALI. Moreover, compared to patients without ALI, patients with TBI showed a clearly enhanced inflammatory response that was closely correlated with the blood glutamate levels. The blood glutamate concentration was also found to be a risk factor (adjusted odds ratio, 2.229; 95% CI, 1.082–2.634) and was a better predictor of TBI-ALI than the Glasgow Coma Scale (GCS) score. These results indicated that dramatically increased blood glutamate concentrations were closely related to the occurrence of TBI-ALI and could be used as a predictive marker for “at-risk” patients.
Collapse
Affiliation(s)
- Wei Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wan-Li Zhu
- Department of Neurosurgery, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ya-Lei Ning
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ping Li
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Zhao
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Nan Yang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xing Chen
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu-Lin Jiang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wen-Qun Yang
- Department of ICU, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Dong-Po Jiang
- Department of ICU, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Li-Yong Chen
- Department of Anesthesiology, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China.
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
30
|
Zhao ZA, Zhao Y, Ning YL, Yang N, Peng Y, Li P, Chen XY, Liu D, Wang H, Chen X, Bai W, Chen JF, Zhou YG. Adenosine A 2A receptor inactivation alleviates early-onset cognitive dysfunction after traumatic brain injury involving an inhibition of tau hyperphosphorylation. Transl Psychiatry 2017; 7:e1123. [PMID: 28485728 PMCID: PMC5534966 DOI: 10.1038/tp.2017.98] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 03/21/2017] [Accepted: 04/04/2017] [Indexed: 12/16/2022] Open
Abstract
Tau is a microtubule-associated protein, and the oligomeric and hyperphosphorylated forms of tau are increased significantly after neurotrauma and considered important factors in mediating cognitive dysfunction. Blockade of adenosine A2A receptors, either by caffeine or gene knockout (KO), alleviates cognitive dysfunction after traumatic brain injury (TBI). We postulated that A2AR activation exacerbates cognitive impairment via promoting tau hyperphosphorylation. Using a mouse model of moderate controlled cortical impact, we showed that TBI induced hyperphosphorylated tau (p-tau) in the hippocampal dentate gyrus and spatial memory deficiency in the Morris water maze test at 7 days and 4 weeks after TBI. Importantly, pharmacological blockade (A2AR antagonist ZM241385 or non-selective adenosine receptor antagonist caffeine) or genetic inactivation of A2ARs reduced the level of tau phosphorylation at Ser404 and alleviated spatial memory dysfunction. The A2AR control of p-tau is further supported by the observations that a KO of A2AR decreased the activity of the tau phosphorylation kinases, glycogen synthase kinase-3β (GSK-3β) and protein kinase A (PKA) after TBI, and by that CGS21680 (A2AR agonist) exacerbated okadaic acid-induced tau hyperphosphorylation in cultured primary hippocampal neurons. Lastly, CGS21680-induced neuronal tau hyperphosphorylation and axonal injury were effectively alleviated by individual treatments with ZM241385 (A2AR antagonist), H89 (PKA antagonist) and SB216763 (GSK-3β antagonist), or by the combined treatment with H89 and SB216763. Our findings suggest a novel mechanism whereby A2AR activation triggers cognitive dysfunction by increasing the phosphorylation level of tau protein after TBI and suggest a promising therapeutic and prophylactic strategy by targeting aberrant A2AR signaling via tau phosphorylation.
Collapse
Affiliation(s)
- Z-A Zhao
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y Zhao
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y-L Ning
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - N Yang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y Peng
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - P Li
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - X-Y Chen
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - D Liu
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - H Wang
- Department of Neurosurgery, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - X Chen
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - W Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - J-F Chen
- Department of Neurology and Pharmacology, Boston University School of Medicine, Boston, MA, USA
| | - Y-G Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China,Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing 400042, China. E-mail: or
| |
Collapse
|
31
|
Zhao ZA, Ning YL, Li P, Yang N, Peng Y, Xiong RP, Zhao Y, Liu D, Zeng XJ, Chen JF, Zhou YG. Widespread hyperphosphorylated tau in the working memory circuit early after cortical impact injury of brain (Original study). Behav Brain Res 2017; 323:146-153. [DOI: 10.1016/j.bbr.2017.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/01/2017] [Indexed: 12/14/2022]
|
32
|
Ning YL, Yang N, Chen X, Zhao ZA, Zhang XZ, Chen XY, Li P, Zhao Y, Zhou YG. Chronic caffeine exposure attenuates blast-induced memory deficit in mice. Chin J Traumatol 2017; 18:204-11. [PMID: 26764541 DOI: 10.1016/j.cjtee.2015.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To investigate the effects of three different ways of chronic caffeine administration on blast- induced memory dysfunction and to explore the underlying mechanisms. METHODS Adult male C57BL/6 mice were used and randomly divided into five groups: control: without blast exposure, con-water: administrated with water continuously before and after blast-induced traumatic brain injury (bTBI), con-caffeine: administrated with caffeine continuously for 1 month before and after bTBI, pre-caffeine: chronically administrated with caffeine for 1 month before bTBI and withdrawal after bTBI, post-caffeine: chronically administrated with caffeine after bTBI. After being subjected to moderate intensity of blast injury, mice were recorded for learning and memory performance using Morris water maze (MWM) paradigms at 1, 4, and 8 weeks post-blast injury. Neurological deficit scoring, glutamate concentration, proinflammatory cytokines production, and neuropathological changes at 24 h, 1, 4, and 8 weeks post-bTBI were examined to evaluate the brain injury in early and prolonged stages. Adenosine A1 receptor expression was detected using qPCR. RESULTS All of the three ways of chronic caffeine exposure ameliorated blast-induced memory deficit, which is correlated with the neuroprotective effects against excitotoxicity, inflammation, astrogliosis and neuronal loss at different stages of injury. Continuous caffeine treatment played positive roles in both early and prolonged stages of bTBI; pre-bTBI and post-bTBI treatment of caffeine tended to exert neuroprotective effects at early and prolonged stages of bTBI respectively. Up-regulation of adenosine A1 receptor expression might contribute to the favorable effects of chronic caffeine consumption. CONCLUSION Since caffeinated beverages are widely consumed in both civilian and military personnel and are convenient to get, the results may provide a promising prophylactic strategy for blast-induced neurotrauma and the consequent cognitive impairment.
Collapse
Affiliation(s)
- Ya-Lei Ning
- Molecular Biology Center, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery and Daping Hospital,Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhao B, Gao WW, Liu YJ, Jiang M, Liu L, Yuan Q, Hou JB, Xia ZY. The role of glycogen synthase kinase 3 beta in brain injury induced by myocardial ischemia/reperfusion injury in a rat model of diabetes mellitus. Neural Regen Res 2017; 12:1632-1639. [PMID: 29171428 PMCID: PMC5696844 DOI: 10.4103/1673-5374.217337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Myocardial ischemia/reperfusion injury can lead to severe brain injury. Glycogen synthase kinase 3 beta is known to be involved in myo-cardial ischemia/reperfusion injury and diabetes mellitus. However, the precise role of glycogen synthase kinase 3 beta in myocardial ischemia/reperfusion injury-induced brain injury is unclear. In this study, we observed the effects of glycogen synthase kinase 3 beta on brain injury induced by myocardial ischemia/reperfusion injury in diabetic rats. Rat models of diabetes mellitus were generated via intraperitoneal injection of streptozotocin. Models of myocardial ischemia/reperfusion injury were generated by occluding the anterior descending branch of the left coronary artery. Post-conditioning comprised three cycles of ischemia/reperfusion. Immunohistochemical staining and western blot assays demonstrated that after 48 hours of reperfusion, the structure of the brain was seriously damaged in the experimental rats compared with normal controls. Expression of Bax, interleukin-6, interleukin-8, terminal deoxynucleotidyl transferase dUTP nick end labeling, and cleaved caspase-3 in the brain was significantly increased, while expression of Bcl-2, interleukin-10, and phospho-glycogen synthase kinase 3 beta was decreased. Diabetes mellitus can aggravate inflammatory reactions and apoptosis. Ischemic post-conditioning with glycogen synthase kinase 3 beta inhibitor lithium chloride can effectively reverse these changes. Our results showed that myocardial ischemic post-conditioning attenuated myocardial ischemia/reperfusion injury-induced brain injury by activating glyco-gen synthase kinase 3 beta. According to these results, glycogen synthase kinase 3 beta appears to be an important factor in brain injury induced by myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Wen-Wei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Ya-Jing Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Meng Jiang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lian Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Quan Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jia-Bao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
34
|
Activation of Adenosine 2A receptor inhibits neutrophil apoptosis in an autophagy-dependent manner in mice with systemic inflammatory response syndrome. Sci Rep 2016; 6:33614. [PMID: 27647162 PMCID: PMC5028892 DOI: 10.1038/srep33614] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/30/2016] [Indexed: 01/13/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is an overwhelming whole body inflammation caused by infectious diseases or sterile insults. Neutrophils are the dominant participants during inflammation, and their survival and death determine the initiation as well as resolution of SIRS. Apoptosis and autophagy are two fundamental cellular processes that modulating cell fate, but their correlation and regulators in neutrophils under SIRS condition have not been elucidated. In this study, we demonstrated that high dose of LPS induced both apoptosis and autophagy of neutrophils in a mouse SIRS model and LPS-stimulated neutrophils in vitro. Moreover, we found that the adenosine 2A receptor (A2AR), a known anti-inflammatory G protein-coupled receptor (GPCR), could inhibit LPS-induced neutrophil apoptosis by suppressing the LPS-induced autophagy. Activation of A2AR suppressed LPS-induced autophagy by inhibiting the ROS-JNK pathway as well as promoting GPCR βϒ subunit–AKT signaling. The A2AR-inhibited autophagy suppressed apoptosis of neutrophils by blocking caspase8, caspase3 and PARP signaling. These findings not only increase our understandings of neutrophils’ fate and function in response to systemic inflammation, but also identify a novel anti-inflammatory role of A2AR in modulating neutrophils’ survival during inflammation.
Collapse
|
35
|
ARDS in the brain-injured patient: what's different? Intensive Care Med 2016; 42:790-793. [PMID: 26969670 DOI: 10.1007/s00134-016-4298-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 02/26/2016] [Indexed: 10/22/2022]
|
36
|
Weber DJ, Gracon ASA, Ripsch MS, Fisher AJ, Cheon BM, Pandya PH, Vittal R, Capitano ML, Kim Y, Allette YM, Riley AA, McCarthy BP, Territo PR, Hutchins GD, Broxmeyer HE, Sandusky GE, White FA, Wilkes DS. The HMGB1-RAGE axis mediates traumatic brain injury-induced pulmonary dysfunction in lung transplantation. Sci Transl Med 2015; 6:252ra124. [PMID: 25186179 DOI: 10.1126/scitranslmed.3009443] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) results in systemic inflammatory responses that affect the lung. This is especially critical in the setting of lung transplantation, where more than half of donor allografts are obtained postmortem from individuals with TBI. The mechanism by which TBI causes pulmonary dysfunction remains unclear but may involve the interaction of high-mobility group box-1 (HMGB1) protein with the receptor for advanced glycation end products (RAGE). To investigate the role of HMGB1 and RAGE in TBI-induced lung dysfunction, RAGE-sufficient (wild-type) or RAGE-deficient (RAGE(-/-)) C57BL/6 mice were subjected to TBI through controlled cortical impact and studied for cardiopulmonary injury. Compared to control animals, TBI induced systemic hypoxia, acute lung injury, pulmonary neutrophilia, and decreased compliance (a measure of the lungs' ability to expand), all of which were attenuated in RAGE(-/-) mice. Neutralizing systemic HMGB1 induced by TBI reversed hypoxia and improved lung compliance. Compared to wild-type donors, lungs from RAGE(-/-) TBI donors did not develop acute lung injury after transplantation. In a study of clinical transplantation, elevated systemic HMGB1 in donors correlated with impaired systemic oxygenation of the donor lung before transplantation and predicted impaired oxygenation after transplantation. These data suggest that the HMGB1-RAGE axis plays a role in the mechanism by which TBI induces lung dysfunction and that targeting this pathway before transplant may improve recipient outcomes after lung transplantation.
Collapse
Affiliation(s)
- Daniel J Weber
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Adam S A Gracon
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S Ripsch
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Amanda J Fisher
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Bo M Cheon
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Pankita H Pandya
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ragini Vittal
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Youngsong Kim
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Yohance M Allette
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - Amanda A Riley
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brian P McCarthy
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul R Territo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gary D Hutchins
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - George E Sandusky
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fletcher A White
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| | - David S Wilkes
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
37
|
Adenosine A2A receptor deficiency alleviates blast-induced cognitive dysfunction. J Cereb Blood Flow Metab 2013; 33:1789-98. [PMID: 23921902 PMCID: PMC3824177 DOI: 10.1038/jcbfm.2013.127] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 07/06/2013] [Accepted: 07/09/2013] [Indexed: 11/09/2022]
Abstract
Traumatic brain injury (TBI), particularly explosive blast-induced TBI (bTBI), has become the most prevalent injury among military personnel. The disruption of cognitive function is one of the most serious consequences of bTBI because its long-lasting effects prevent survivors fulfilling their active duty and resuming normal civilian life. However, the mechanisms are poorly understood and there is no treatment available. This study investigated the effects of adenosine A2A receptor (A2AR) on bTBI-induced cognitive deficit, and explored the underlying mechanisms. After being subjected to moderate whole-body blast injury, mice lacking the A2AR (A2AR knockout (KO)) showed less severity and shorter duration of impaired spatial reference memory and working memory than wild-type mice did. In addition, bTBI-induced cortical and hippocampal lesions, as well as proinflammatory cytokine expression, glutamate release, edema, cell loss, and gliosis in both early and prolonged phases of the injury, were significantly attenuated in A2AR KO mice. The results suggest that early injury and chronic neuropathological damages are important mechanisms of bTBI-induced cognitive impairment, and that the impairment can be attenuated by preventing A2AR activation. These findings suggest that A2AR antagonism is a potential therapeutic strategy for mild-to-moderate bTBI and consequent cognitive impairment.
Collapse
|