1
|
Wang R, Gan C, Gong B, Huang J, Lou Z, Wang D, Yan R, Li G, Xiong T, Guo J. Tongfu Xingshen capsule alleviates stroke-associated pneumonia-induced multiple organ injuries by modulating the gut microbiota and sphingolipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156756. [PMID: 40252432 DOI: 10.1016/j.phymed.2025.156756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/14/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Stroke-associated pneumonia (SAP) represents a major complication and cause of death in patients suffering from intracerebral haemorrhage (ICH). It's urgent to develop more effective therapeutic strategies. Tongfu Xingshen capsule (TFXS) is a traditional Chinese medicine that has been utilised in clinical studies for the treatment of ICH and SAP, but the underlying mechanisms remain to be fully elucidated. PURPOSE This study aims to explore the therapeutic effects and mechanisms of TFXS on SAP using an aspiration-induced Klebsiella pneumoniae infection-complicating ICH rat model and an intratracheal injection of lipopolysaccharide (LPS)-induced acute lung injury-complicating ICH rat model. METHODS The chemical components of TFXS are characterised using ULPLC-Q Exactive-Orbitrap-MS. The therapeutic effects of TFXS are evaluated through neurological scoring, histopathology analysis, magnetic resonance imaging, immunofluorescence, Alcian blue-nuclear fast red staining, myeloperoxidase activity assessment, leukocyte counting, and ELISA. To investigate the underlying mechanisms, faecal microbiota transplantation, 16S rRNA sequencing, untargeted metabolomics, and Spearman correlation analyses are performed. RESULTS A total of 60 compounds are identified in TFXS. Pharmacological analysis reveals that TFXS significantly mitigates neurological deficits, enhances haematoma absorption, attenuates brain damage and neuroinflammation, and improves pneumonia and pulmonary injury by reducing the infiltration of leukocytes and lymphocytes, as well as suppressing the infiltration and overactivation of neutrophils. TFXS also alleviates intestinal lesions and barrier damage by increasing acidic mucins and the expression of the tight junction protein zonula occludens-1 (ZO-1). Mechanistically, TFXS ameliorates pneumonia and pulmonary injury in a gut microbiota-dependent manner. It reverses sphingolipid metabolism disorders and ceramide accumulation by modulating SAP-induced gut microbiota dysbiosis and enhancing the abundance of probiotics, including Lactobacillus, Allobaculum and Enterococcus. CONCLUSION TFXS exerts anti-inflammatory and protective effects on the brain, lung, and gut by alleviating gut microbiota dysbiosis and sphingolipid metabolism disorders. These findings highlight TFXS as a promising therapeutic candidate for the treatment of SAP.
Collapse
Affiliation(s)
- Ruihua Wang
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Changlian Gan
- School of Traditional Dai Medicine, West Yunnan University of Applied Science, JH, Xishuangbanna, Yunnan Province, 666100, PR China
| | - Baoying Gong
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Juan Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Zhenzhen Lou
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Daxiu Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, PR China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510000, PR China.
| | - Tianqin Xiong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 511400, PR China.
| | - Jianwen Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Neurology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China.
| |
Collapse
|
2
|
Wang R, Gan C, Mao R, Chen Y, Yan R, Li G, Xiong T, Guo J. Rat models of postintracerebral hemorrhage pneumonia induced by nasal inoculation with Klebsiella pneumoniae or intratracheal inoculation with LPS. Front Immunol 2025; 15:1477902. [PMID: 39845950 PMCID: PMC11750689 DOI: 10.3389/fimmu.2024.1477902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background A stable and reproducible experimental bacterial pneumonia model postintracerebral hemorrhage (ICH) is necessary to help investigating the pathogenesis and novel treatments of Stroke-associated pneumonia (SAP). Aim To establish a Gram-negative bacterial pneumonia-complicating ICH rat model and an acute lung injury (ALI)-complicating ICH rat model. Methods We established two standardized models of post-ICH pneumonia by nasal inoculation with Klebsiella pneumoniae (Kp) or intratracheal inoculation with lipopolysaccharide (LPS). Survival and neurological scores were monitored. Magnetic resonance imaging was performed to evaluate hematoma volume. Abdominal aortic blood was collected for leukocyte counting, serum was isolated to determine concentrations of S100β and proinflammatory cytokines using ELISAs. Histopathological changes of brain, lung and gut were assessed using hematoxylin-eosin staining. Lung was isolated for immunofluorescence staining for myeloperoxidase (MPO). Bronchoalveolar lavage fluid was collected for leukocyte counting, and supernatant was prepared to measure MPO activity. Ileum was isolated for immunofluorescence staining for tight junction proteins ZO-1 and γδ TCRs/IL-17A and for Alcian blue-nuclear fast red staining of acidic mucins. Feces were collected, 16S rRNA sequencing, untargeted metabolomics and Spearman's correlation analyses were performed to explore changes of gut microbiota, metabolites and their interactions. Results In Kp-induced bacterial pneumonia-complicating ICH rats, we demonstrated that Kp challenge caused more severe neurological deficits, brain damage, neuroinflammation, and aggravated pneumonia and lung injury. Disruptions of the intestinal structure and gut barrier and the reductions of the protective intestinal IL-17A-producing γδT cells were also observed. Kp challenge exacerbated the gut microbiota dysbiosis and fecal metabolic profile disorders, which were characterized by abnormal sphingolipid metabolism especially elevated ceramide levels; increased levels of neurotoxic quinolinic acid and an upregulation of tryptophan (Trp)-serotonin-melatonin pathway. Spearman's correlation analyses further revealed that the reduction or depletion of some beneficial bacteria, such as Allobaculum and Faecalitalea, and the blooming of some opportunistic pathogens, such as Turicibacter, Dietzia, Corynebacterium and Clostridium_sensu_stricto_1 in Kp-induced SAP rats were associated with the disordered sphingolipid and Trp metabolism. Using an LPS-induced ALI complicating ICH model, we also characterized SAP-induced brain, lung and gut histopathology injuries; peripheral immune disorders and intense pulmonary inflammatory responses. Conclusions These two models may be highly useful for investigating the pathogenesis and screening and optimizing potential treatments for SAP. Moreover, the differential genera and sphingolipid or Trp metabolites identified above seem to be promising therapeutic targets.
Collapse
Affiliation(s)
- Ruihua Wang
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changlian Gan
- School of Traditional Dai Medicine, West Yunnan University of Applied Science, Xishuangbanna, China
| | - Rui Mao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Chen
- Department of Bioinformatics, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianqin Xiong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Neurology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Joaquim LS, Steiner B, Farias B, Machado RS, Danielski LG, Mathias K, Stork S, Lanzzarin E, Novaes L, Bonfante S, Generoso JDS, Alano CG, Lemos I, Dominguini D, Giustina AD, Catalão CHR, Streck EL, Giridharan VV, Dal-Pizzol F, Barichello T, de Bitencourt RM, Petronilho F. Sepsis compromises post-ischemic stroke neurological recovery and is associated with sex differences. Life Sci 2024; 349:122721. [PMID: 38754813 DOI: 10.1016/j.lfs.2024.122721] [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: 03/08/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
AIMS Infection is a complication after stroke and outcomes vary by sex. Thus, we investigated if sepsis affects brain from ischemic stroke and sex involvement. MAIN METHODS Male and female Wistar rats, were submitted to middle cerebral artery occlusion (MCAO) and after 7 days sepsis to cecal ligation and perforation (CLP). Infarct size, neuroinflammation, oxidative stress, and mitochondrial activity were quantified 24 h after CLP in the prefrontal cortex and hippocampus. Survival and neurological score were assessed up to 15 days after MCAO or 8 days after CLP (starting at 2 h after MCAO) and memory at the end. KEY FINDINGS CLP decreased survival, increased neurological impairments in MCAO females. Early, in male sepsis following MCAO led to increased glial activation in the brain structures, and increased TNF-α and IL-1β in the hippocampus. All groups had higher IL-6 in both tissues, but the hippocampus had lower IL-10. CLP potentiated myeloperoxidase (MPO) in the prefrontal cortex of MCAO male and female. In MCAO+CLP, only male increased MPO and nitrite/nitrate in hippocampus. Males in all groups had protein oxidation in the prefrontal cortex, but only MCAO+CLP in the hippocampus. Catalase decreased in the prefrontal cortex and hippocampus of all males and females, and MCAO+CLP only increased this activity in males. Female MCAO+CLP had higher prefrontal cortex complex activity than males. In MCAO+CLP-induced long-term memory impairment only in females. SIGNIFICANCE The parameters evaluated for early sepsis after ischemic stroke show a worse outcome for males, while females are affected during long-term follow-up.
Collapse
Affiliation(s)
- Larissa Silva Joaquim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Beatriz Steiner
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Brenno Farias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Richard Simon Machado
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Lucineia Gainski Danielski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil; Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Khiany Mathias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Solange Stork
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Everton Lanzzarin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Linerio Novaes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Sandra Bonfante
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Jaqueline da Silva Generoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Carolina Giassi Alano
- Laboratory of Experimental Biomedicine, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Isabela Lemos
- Laboratory of Experimental Biomedicine, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Amanda Della Giustina
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, ON, Canada
| | - Carlos Henrique Rocha Catalão
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Emilio Luiz Streck
- Laboratory of Experimental Biomedicine, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Vijayasree V Giridharan
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil; Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Rafael Mariano de Bitencourt
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil.
| |
Collapse
|
4
|
Wang Y, Liu C, Ren Y, Song J, Fan K, Gao L, Ji X, Chen X, Zhao H. Nanomaterial-Based Strategies for Attenuating T-Cell-Mediated Immunodepression in Stroke Patients: Advancing Research Perspectives. Int J Nanomedicine 2024; 19:5793-5812. [PMID: 38882535 PMCID: PMC11180442 DOI: 10.2147/ijn.s456632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
This review article discusses the potential of nanomaterials in targeted therapy and immunomodulation for stroke-induced immunosuppression. Although nanomaterials have been extensively studied in various biomedical applications, their specific use in studying and addressing immunosuppression after stroke remains limited. Stroke-induced neuroinflammation is characterized by T-cell-mediated immunodepression, which leads to increased morbidity and mortality. Key observations related to immunodepression after stroke, including lymphopenia, T-cell dysfunction, regulatory T-cell imbalance, and cytokine dysregulation, are discussed. Nanomaterials, such as liposomes, micelles, polymeric nanoparticles, and dendrimers, offer advantages in the precise delivery of drugs to T cells, enabling enhanced targeting and controlled release of immunomodulatory agents. These nanomaterials have the potential to modulate T-cell function, promote neuroregeneration, and restore immune responses, providing new avenues for stroke treatment. However, challenges related to biocompatibility, stability, scalability, and clinical translation need to be addressed. Future research efforts should focus on comprehensive studies to validate the efficacy and safety of nanomaterial-based interventions targeting T cells in stroke-induced immunosuppression. Collaborative interdisciplinary approaches are necessary to advance the field and translate these innovative strategies into clinical practice, ultimately improving stroke outcomes and patient care.
Collapse
Grants
- This work was supported by the National Natural Science Foundation of China (Grant number 82001248), National University of Singapore (NUHSRO/2020/133/Startup/08, NUHSRO/2023/008/NUSMed/TCE/LOA, NUHSRO/2021/034/TRP/09/Nanomedicine, NUHSRO/2021/044/Kickstart/09/LOA, 23-0173-A0001), National Medical Research Council (MOH-001388-00, CG21APR1005, OFIRG23jul-0047), Singapore Ministry of Education (MOE-000387-00), and National Research Foundation (NRF-000352-00)
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Yanhong Ren
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Zhang J, Ling L, Xiang L, Li W, Bao P, Yue W. Role of the gut microbiota in complications after ischemic stroke. Front Cell Infect Microbiol 2024; 14:1334581. [PMID: 38644963 PMCID: PMC11026644 DOI: 10.3389/fcimb.2024.1334581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Ischemic stroke (IS) is a serious central nervous system disease. Post-IS complications, such as post-stroke cognitive impairment (PSCI), post-stroke depression (PSD), hemorrhagic transformation (HT), gastrointestinal dysfunction, cardiovascular events, and post-stroke infection (PSI), result in neurological deficits. The microbiota-gut-brain axis (MGBA) facilitates bidirectional signal transduction and communication between the intestines and the brain. Recent studies have reported alterations in gut microbiota diversity post-IS, suggesting the involvement of gut microbiota in post-IS complications through various mechanisms such as bacterial translocation, immune regulation, and production of gut bacterial metabolites, thereby affecting disease prognosis. In this review, to provide insights into the prevention and treatment of post-IS complications and improvement of the long-term prognosis of IS, we summarize the interaction between the gut microbiota and IS, along with the effects of the gut microbiota on post-IS complications.
Collapse
Affiliation(s)
- Jinwei Zhang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Ling Ling
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Lei Xiang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Wenxia Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Pengnan Bao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Wei Yue
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
6
|
Miao Y, Fan X, Wei L, Wang B, Diao F, Fu J, Zhuang P, Zhang Y. Lizhong decoction ameliorates pulmonary infection secondary to severe traumatic brain injury in rats by regulating the intestinal physical barrier and immune response. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116346. [PMID: 36898448 DOI: 10.1016/j.jep.2023.116346] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The pathogenesis of pulmonary infection secondary to severe traumatic brain injury (sTBI) is closely related to damage to the intestinal barrier. Lizhong decoction (LZD) is a prominent traditional Chinese medicine (TCM) that is widely used in clinical treatment to regulate gastrointestinal movement and enhance resistance. Nevertheless, the role and mechanism of LZD in lung infection secondary to sTBI have yet to be elucidated. AIM OF THE STUDY Here, we evaluate the therapeutic effect of LZD on pulmonary infection secondary to sTBI in rats and discuss potential regulatory mechanisms. MATERIALS AND METHODS The chemical constituents of LZD were analyzed by ultra-high performance liquid chromatography-Q Exactive-tandem mass spectrometry(UPLC-QE-MS/MS). The efficacy of LZD on rats with lung infection secondary to sTBI was examined by changes in brain morphology, coma time, brain water content, mNSS score, colony counts, 16S rRNA/RNaseP/MRP30 kDa(16S/RPP30), myeloperoxidase (MPO) content and pathology of lung tissue. The concentration of fluorescein isothiocyanate(FITC)-dextran in serum and the contents of secretory immunoglobulin A (SIgA) in colon tissue were detected by enzyme-linked immunosorbent assay (ELISA). Subsequently, Alcian Blue Periodic acid Schiff (AB-PAS) was used to detect colonic goblet cells. Immunofluorescence (IF) was used to detect the expression of tight junction proteins. The proportions of CD3+ cell, CD4+CD8+ T cells, CD45+ cell and CD103+ cells in the colon were analyzed by flow cytometry (FC). In addition, colon transcriptomics were analyzed by Illumina mRNA-Seq sequencing. Real-time quantitative polymerase chain reaction (qRT‒PCR) was used to verify the genes associated with LZD alleviation of intestinal barrier function. RESULTS Twenty-nine chemical constituents of LZD were revealed with UPLC-QE-MS/MS analysis. Administration of LZD significantly reduced colony counts, 16S/RPP30 and MPO content in lung infection secondary to sTBI rats. In addition, LZD also reduced the serum FITC-glucan content and the SIgA content of the colon. Additionally, LZD significantly increased the number of colonic goblet cells and the expression of tight junction proteins. Furthermore, LZD significantly decreased the proportion of CD3+ cell, CD4+CD8+ T cells,CD45+ and CD103+ cells in colon tissue. Transcriptomic analysis identified 22 upregulated genes and 56 downregulated genes in sTBI compared to the sham group. The levels of seven genes were recovered after LZD treatment. qRT‒PCR successfully validated two genes (Jchain and IL-6) at the mRNA level. CONCLUSION LZD can improves sTBI secondary lung infection by regulating the intestinal physical barrier and immune response. Thees results suggested that LZD may be a prospective treatment for pulmonary infection secondary to sTBI.
Collapse
Affiliation(s)
- Yulu Miao
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuejin Fan
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luge Wei
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Wang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fengyin Diao
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiafeng Fu
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengwei Zhuang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Yanjun Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
7
|
Sung LC, Chang CC, Yeh CC, Lee CY, Hu CJ, Cherng YG, Chen TL, Liao CC. The effects of regular dental scaling on the complications and mortality after stroke: a retrospective cohort study based on a real-world database. BMC Oral Health 2023; 23:487. [PMID: 37452324 PMCID: PMC10349500 DOI: 10.1186/s12903-023-03178-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/27/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Previous observational studies have shown that people with dental scaling (DS) had decreased risk of stroke. However, limited information is available on the association between DS and poststroke outcomes. The present study aimed to evaluate the effects of regular DS on the complications and mortality after stroke. METHODS We conducted a retrospective cohort study of 49,547 hospitalized stroke patients who received regular DS using 2010-2017 claims data of Taiwan's National Health Insurance. Using a propensity-score matching procedure, we selected 49,547 women without DS for comparison. Multiple logistic regressions were used to calculate the odds ratios (ORs) and 95% confidence intervals (CIs) of poststroke complications and in-hospital mortality associated with regular DS. RESULTS Stroke patients with regular DS had significantly lower risks of poststroke pneumonia (OR 0.58, 95% CI 0.54-0.63), septicemia (OR 0.58, 95% CI 0.54-0.63), urinary tract infection (OR 0.68, 95% CI 0.66-0.71), intensive care (OR 0.81, 95% CI 0.78-0.84), and in-hospital mortality (OR 0.66, 95% CI 0.62-0.71) compared with non-DS stroke patients. Stroke patients with regular DS also had shorter hospital stays (p < 0.0001) and less medical expenditures (p < 0.0001) during stroke admission than the control group. Lower rates of poststroke adverse events in patients with regular DS were noted in both sexes, all age groups, and people with various types of stroke. CONCLUSION Stroke patients with regular DS showed fewer complications and lower mortality compared with patients had no DS. These findings suggest the urgent need to promote regular DS for this susceptible population of stroke patients.
Collapse
Affiliation(s)
- Li-Chin Sung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Chuen-Chau Chang
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Surgery, University of Illinois, Chicago, United States of America
| | - Chia-Yen Lee
- Department of Dentistry, Taiwan Adventist Hospital, Taipei, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yih-Giun Cherng
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ta-Liang Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
8
|
Han S, Cai L, Chen P, Kuang W. A study of the correlation between stroke and gut microbiota over the last 20years: a bibliometric analysis. Front Microbiol 2023; 14:1191758. [PMID: 37350780 PMCID: PMC10282156 DOI: 10.3389/fmicb.2023.1191758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Purpose This study intends to uncover a more thorough knowledge structure, research hotspots, and future trends in the field by presenting an overview of the relationship between stroke and gut microbiota in the past two decades. Method Studies on stroke and gut microbiota correlations published between 1st January 2002 and 31st December 2021 were retrieved from the Web of Science Core Collection and then visualized and scientometrically analyzed using CiteSpace V. Results A total of 660 papers were included in the study, among which the United States, the United Kingdom, and Germany were the leading research centers. Cleveland Clinic, Southern Medical University, and Chinese Academy of Science were the top three institutions. The NATURE was the most frequently co-cited journal. STANLEY L HAZEN was the most published author, and Tang WHW was the most cited one. The co-occurrence analysis revealed eight clusters (i.e., brain-gut microbiota axis, fecal microbiome transplantation, gut microbiota, hypertension, TMAO, ischemic stroke, neuroinflammation, atopobiosis). "gut microbiota," "Escherichia coli," "cardiovascular disease," "risk," "disease," "ischemic stroke," "stroke," "metabolism," "inflammation," and "phosphatidylcholine" were the most recent keyword explosions. Conclusion Findings suggest that in the next 10 years, the number of publications produced annually may increase significantly. Future research trends tend to concentrate on the mechanisms of stroke and gut microbiota, with the inflammation and immunological mechanisms, TMAO, and fecal transplantation as hotspots. And the relationship between these mechanisms and a particular cardiovascular illness may also be a future research trend.
Collapse
Affiliation(s)
- Shengnan Han
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Longhui Cai
- First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peipei Chen
- School of Medical Technology, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
9
|
Zhang S, Jin M, Ren J, Sun X, Zhang Z, Luo Y, Sun X. New insight into gut microbiota and their metabolites in ischemic stroke: A promising therapeutic target. Biomed Pharmacother 2023; 162:114559. [PMID: 36989717 DOI: 10.1016/j.biopha.2023.114559] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
The gut-brain axis has been shown to play a vital role in the prognosis and recovery of ischemic stroke (IS), which is associated with gut microbiota dysfunction and changes in the gastrointestinal system and epithelial barrier integrity. In turn, gut microbiota and its derived metabolites can influence stroke outcomes. In this review, we first describe the relationship between IS (clinical and experimental IS) and the gut microbiota. Second, we summarize the role and specific mechanisms of microbiota-derived metabolites in IS. Further, we discuss the roles of natural medicines targeting the gut microbiota. Finally, the potential use of the gut microbiota and derived metabolites as a promising therapeutic opportunity for stroke prevention, diagnosis, and treatment is explored.
Collapse
|
10
|
Premorbid Use of Beta-Blockers or Angiotensin-Converting Enzyme Inhibitors/Angiotensin Receptor Blockers in Patients with Acute Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7733857. [PMID: 36778208 PMCID: PMC9908343 DOI: 10.1155/2023/7733857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/07/2022] [Accepted: 10/14/2022] [Indexed: 02/05/2023]
Abstract
This study was designed to investigate the impact of the preexisting use of beta-blockers, angiotensin-converting enzyme inhibitors (ACEIs), or angiotensin receptor blockers (ARBs) on the cellular immune response in peripheral blood and the clinical outcomes of patients with acute ischemic stroke. We retrospectively collected clinical data from a cohort of 69 patients with premorbid beta-blockers and 56 patients with premorbid ACEIs/ARBs. Additionally, we selected a cohort of 107 patients with acute ischemic stroke to be the control of the same age and sex. We analyzed cellular immune parameters in peripheral blood 1 day after the appearance of symptoms, including the frequencies of circulating white blood cell subpopulations, the neutrophil-to-lymphocyte ratio (NLR), and the lymphocyte-to-monocyte ratio (LMR). We found that the count of lymphocytes and the lymphocyte-to-monocyte ratio were significantly higher in the peripheral blood of patients treated with beta-blockers before stroke than in matched controls. However, the premorbid use of ACEIs/ARBs did not considerably impact the circulating immune parameters listed above in patients with acute ischemic stroke. Furthermore, we found that premorbid use of beta-blockers or ACEIs/ARBs did not significantly change functional outcomes in patients 3 months after the onset of stroke. These results suggest that premorbid use of beta-blockers, but not ACEIs/ARBs, reversed lymphopenia associated with acute ischemic stroke. As cellular immune changes in peripheral blood could be an independent predictor of stroke prognosis, more large-scale studies are warranted to further verify the impact of premorbid use of beta-blockers or ACEIs/ARBs on the prognosis of patients with ischemic stroke. Our research is beneficial to understanding the mechanism of the systemic immune response induced by stroke and has the potential for a therapeutic strategy in stroke interventions and treatment.
Collapse
|
11
|
Gerganova G, Riddell A, Miller AA. CNS border-associated macrophages in the homeostatic and ischaemic brain. Pharmacol Ther 2022; 240:108220. [PMID: 35667516 DOI: 10.1016/j.pharmthera.2022.108220] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022]
Abstract
CNS border-associated macrophages (BAMs) are a small population of specialised macrophages localised in the choroid plexus, meningeal and perivascular spaces. Until recently, the function of this elusive cell type was poorly understood and largely overlooked, especially in comparison to microglia, the primary brain resident immune cell. However, the recent single cell immunophenotyping or transcriptomic analysis of BAM subsets in the homeostatic brain, coupled with the rapid emergence of new studies exploring BAM functions in various cerebral pathologies, including Alzheimer's disease, hypertension-induced neurovascular and cognitive dysfunction, and ischaemic stroke, has unveiled previously unrecognised heterogeneity and spatial-temporal complexity in BAM populations as well as their contributions to brain homeostasis and disease. In this review, we discuss the implications of this new-found knowledge on our current understanding of BAM function in ischaemic stroke. We first provide a comprehensive overview and discussion of the cell-surface expression profiles, transcriptional signatures and potential functional phenotypes of homeostatic BAM subsets described in recent studies. Evidence for their putative physiological roles is examined, including their involvement in immunological surveillance, waste clearance, and vascular permeability. We discuss the evidence supporting the accumulation and genetic transformation of BAMs in response to ischaemia and appraise the experimental evidence that BAM function might be deleterious in the acute phase of stroke, while considering the mechanisms by which BAMs may influence stroke outcomes in the longer term. Finally, we review the therapeutic potential of immunomodulatory strategies as an approach to stroke management, highlighting current challenges in the field and key issues relating to BAMs, and how BAMs could be harnessed experimentally to support future translational research.
Collapse
Affiliation(s)
- Gabriela Gerganova
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Alexandra Riddell
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Alyson A Miller
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom.
| |
Collapse
|
12
|
Wei M, Huang Q, Yu F, Feng X, Luo Y, Zhao T, Tu R, Liao D, Du Y, Huang Q, Gu W, Liu Y, Jiang Y, Gu H, Li Z, Xia J. Stroke-associated infection in patients with co-morbid diabetes mellitus is associated with in-hospital mortality. Front Aging Neurosci 2022; 14:1024496. [PMID: 36533168 PMCID: PMC9751873 DOI: 10.3389/fnagi.2022.1024496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/02/2022] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND AND OBJECTIVE The association between infection and acute ischemic stroke (AIS) with diabetes mellitus (DM) remains unknown. Therefore, this study aimed to explore the effect of infection on AIS with DM. MATERIALS AND METHODS The data of patients with AIS and DM were extracted from the Chinese Stroke Center Alliance (CSCA) database from August 2015 to July 2019. The association between infections [pneumonia or urinary tract infection (UTI)] and in-hospital mortality was analyzed. Logistic regression models were used to identify the risk factors for in-hospital mortality of patients with infection. RESULTS In total, 1,77,923 AIS patients with DM were included in the study. The infection rate during hospitalization was 10.5%, and the mortality rate of infected patients was 3.4%. Stroke-associated infection was an independent risk factor for an early poor functional outcome [odds ratio (OR) = 2.26, 95% confidence interval (CI): 1.97-2.34, P < 0.0001] and in-hospital mortality in AIS patients with DM. The in-hospital mortality after infection was associated with age (OR = 1.02, 95% CI: 1.01-1.03, P < 0.0001), male (OR = 1.39, 95% CI: 1.13-1.71, P = 0.0018), reperfusion therapy (OR = 2.00, 95% CI: 1.56-2.56, P < 0.0001), and fasting plasma glucose at admission (OR = 1.05, 95% CI: 1.03-1.08, P < 0.0001). In contrast, antiplatelet drug therapy (OR = 0.63, 95% CI: 0.50-0.78, P < 0.0001) and hospital stay (OR = 0.96, 95% CI: 0.94-0.97, P < 0.0001) were independent protecting factors against in-hospital mortality of patients with infection. CONCLUSION Infection is an independent risk factor of in-hospital mortality for patients with AIS and DM, and those patients require strengthening nursing management to prevent infection.
Collapse
Affiliation(s)
- Minping Wei
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Yu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xianjing Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yunfang Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Ruxin Tu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Di Liao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qing Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yunhai Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingyu Jiang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongqiu Gu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Li T, Liesz A. Immunity in Stroke: The Next Frontier. Thromb Haemost 2022; 122:1454-1460. [PMID: 35688450 DOI: 10.1055/s-0042-1748890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Translational stroke research has long been focusing on neuroprotective strategies to prevent secondary tissue injury and promote recovery after acute ischemic brain injury. The inflammatory response to stroke has more recently emerged as a key pathophysiological pathway contributing to stroke outcome. It is now accepted that the inflammatory response is functionally involved in all phases of the ischemic stroke pathophysiology. The immune response is therefore considered a breakthrough target for ischemic stroke treatment. On one side, stroke induces a local neuroinflammatory response, in which the inflammatory activation of glial, endothelial and brain-invading cells contributes to lesion progression after stroke. On the other side, ischemic brain injury perturbs systemic immune homeostasis and results in long-lasting changes of systemic immunity. Here, we briefly summarize current concepts in local neuroinflammation and the systemic immune responses after stroke, and highlight two promising therapeutic strategies for poststroke inflammation.
Collapse
Affiliation(s)
- Ting Li
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
14
|
Yang L, Wenping X, Jinfeng Z, Jiangxia P, Jingbo W, Baojun W. Are beta blockers effective in preventing stroke associated infections? - a systematic review and meta-analysis. Aging (Albany NY) 2022; 14:4459-4470. [PMID: 35585021 PMCID: PMC9186777 DOI: 10.18632/aging.204086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/07/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Excessive sympathoexcitation could lead to stroke associated infection. Inhibiting sympathetic excitation may reduce the infection risk after stroke. Thus, the present study aimed to determine the protective effect of beta blockers on stroke associated infection through systematic review and meta-analysis. METHODS A systematic search of multiple databases were performed up to February 2022. The included studies required beta blockers therapy in stroke patients and assessed the incidence of stroke-associated infections. Outcomes of interest included infections, pneumonia, urinary tract infection and sepsis. Random-effects model was used for analysis. Heterogeneity was evaluated using I2 statistics and publication bias was evaluated by the funnel plot. RESULT A total of 83 potentially relevant publications was identified in the initial search. Six studies met the inclusion criteria for meta-analysis. The risk of bias in the included articles satisfies the quality requirement of meta-analysis. No significant associations between beta blockers therapy and the prevention of stroke associated infection, stroke associated pneumonia and septicemia were found, However, subgroup analyses revealed an association between beta blockers treatment and the increased risk of post-stroke urinary tract infection or stroke associated pneumonia in some stroke patients (OR = 1.69 [1.33, 2.14], P < 0.0001; OR = 1.85 [1.51, 2.26], P < 0.0001). CONCLUSION Due to the lack of robust evidence, this meta-analysis may not support the preventive effect of beta blockers on stroke associated infection. But beta blockers treatment may be associated with development of post-stroke urinary tract infection and stroke associated pneumonia in some stroke patients.
Collapse
Affiliation(s)
- Li Yang
- Department of Neurology, Baotou Center Hospital, Inner Mongolia, Baotou, China.,School of Medicine, Inner Mongolia Medical University, Inner Mongolia, Hohhot, China
| | - Xiang Wenping
- Department of Neurology, Baotou Center Hospital, Inner Mongolia, Baotou, China
| | - Zhang Jinfeng
- Department of Neurology, Baotou Center Hospital, Inner Mongolia, Baotou, China
| | - Pang Jiangxia
- Department of Neurology, Baotou Center Hospital, Inner Mongolia, Baotou, China
| | - Wang Jingbo
- Department of Neurology, Baotou Center Hospital, Inner Mongolia, Baotou, China
| | - Wang Baojun
- Department of Neurology, Baotou Center Hospital, Inner Mongolia, Baotou, China
| |
Collapse
|
15
|
Li M, He Z, Yang J, Guo Q, Weng H, Luo J, Gong B, Cui W, Ding B, Guo J. Clinical Characteristics, Outcomes, and Risk Factors of Disease Severity in Patients With COVID-19 and With a History of Cerebrovascular Disease in Wuhan, China: A Retrospective Study. Front Neurol 2022; 12:706478. [PMID: 35087460 PMCID: PMC8787151 DOI: 10.3389/fneur.2021.706478] [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: 05/07/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background and Purpose: Coronavirus disease 2019 (COVID-19) rapidly resulted in a pandemic. Information on patients with a history of cerebrovascular disease (CVD) infected with severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) is limited. This study investigated the clinical features and the risk factors of developing adverse outcomes in patients with COVID-19 and with previous CVD. Methods: This was a single-center retrospective clinical study including all the confirmed cases of COVID-19 at Wuhan Huoshenshan Hospital from February 4 to April 7, 2020. Differences in clinical characteristics were compared between patients with and without a history of CVD. The incidences of severe events comprising all-cause death, intensive care unit admission, shock, and mechanical ventilation usage during hospitalization in two groups were compared using propensity score matching analysis and multivariate logistic regression analyses. Besides, the risk factors of developing severe events in patients with COVID-19 who also have history of CVD were analyzed. Results: A total of 2,554 consecutive patients were included in our study, of whom 109 (4.27%) had a medical history of CVD. Patients with CVD tend to be older and with more comorbidities, including hypertension, diabetes, coronary heart disease, and chronic obstructive pulmonary disease. The levels of white blood cell, neutrophil, C-reactive protein, creatine kinase isoenzymes, and lactate dehydrogenase were higher, whereas the levels of lymphocyte and albumin were lower in the CVD group. Compared to those without CVD, patients with CVD were more likely to have severe events after age matching (12.8 vs. 5.7%, P = 0.012). After adjusting for the confounding effects of age, sex, smoking, and comorbidities, the odds ratio for developing severe events with a history of CVD was 2.326 (95% CI, 1.168–4.630; P = 0.016). Besides, patients with CVD, either with decreased lymphocyte count (OR 9.192, 95% CI, 1.410–59.902, P = 0.020) or increased blood urea nitrogen (OR 5.916, 95% CI, 1.072–32.641, P = 0.041), had a higher risk of developing severe events during hospitalization. Conclusions: Patients with CVD history tend to have adverse clinical outcomes after being infected with SARS-COV-2. Decreased lymphocyte counts and increased blood urea nitrogen levels may be risk factors for adverse outcomes in patients with COVID-19, and had CVD.
Collapse
Affiliation(s)
- Mengzhen Li
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehui He
- Department of Clinical Epidemiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiecong Yang
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qihua Guo
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Heng Weng
- Department of Big Medical Data, Health Construction Administration Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jielian Luo
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baoying Gong
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanzhen Cui
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Banghan Ding
- Department of Emergency, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Mastorakos P, Russo MV, Zhou T, Johnson K, McGavern DB. Antimicrobial immunity impedes CNS vascular repair following brain injury. Nat Immunol 2021; 22:1280-1293. [PMID: 34556874 PMCID: PMC8488012 DOI: 10.1038/s41590-021-01012-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) and cerebrovascular injury are leading causes of disability and mortality worldwide. Systemic infections often accompany these disorders and can worsen outcomes. Recovery after brain injury depends on innate immunity, but the effect of infections on this process is not well understood. Here, we demonstrate that systemically introduced microorganisms and microbial products interfered with meningeal vascular repair after TBI in a type I interferon (IFN-I)-dependent manner, with sequential infections promoting chronic disrepair. Mechanistically, we discovered that MDA5-dependent detection of an arenavirus encountered after TBI disrupted pro-angiogenic myeloid cell programming via induction of IFN-I signaling. Systemic viral infection similarly blocked restorative angiogenesis in the brain parenchyma after intracranial hemorrhage, leading to chronic IFN-I signaling, blood-brain barrier leakage and a failure to restore cognitive-motor function. Our findings reveal a common immunological mechanism by which systemic infections deviate reparative programming after central nervous system injury and offer a new therapeutic target to improve recovery.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Surgical Neurology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Matthew V Russo
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tianzan Zhou
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kory Johnson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
17
|
Effects of CNS Injury-Induced Immunosuppression on Pulmonary Immunity. Life (Basel) 2021; 11:life11060576. [PMID: 34207063 PMCID: PMC8235795 DOI: 10.3390/life11060576] [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: 05/12/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/17/2022] Open
Abstract
Patients suffering from stroke, traumatic brain injury, or other forms of central nervous system (CNS) injury have an increased risk of nosocomial infections due to CNS injury-induced immunosuppression (CIDS). Immediately after CNS-injury, the response in the brain is pro-inflammatory; however, subsequently, local and systemic immunity is suppressed due to the compensatory release of immunomodulatory neurotransmitters. CIDS makes patients susceptible to contracting infections, among which pneumonia is very common and often lethal. Ventilator-acquired pneumonia has a mortality of 20–50% and poses a significant risk to vulnerable patients such as stroke survivors. The mechanisms involved in CIDS are not well understood. In this review, we consolidate the evidence for cellular processes underlying the pathogenesis of CIDS, the emerging treatments, and speculate further on the immune elements at play.
Collapse
|
18
|
Bacterial Pneumonia in Brain-Dead Patients: Clinical Features and Impact on Lung Suitability for Donation. Crit Care Med 2021; 48:1760-1770. [PMID: 33009101 DOI: 10.1097/ccm.0000000000004631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To appraise the epidemiological features of bacterial pneumonia and its impact on lung suitability for donation in brain-dead patients managed with protective ventilatory settings. DESIGN Retrospective observational study. SETTING Six ICUs from two university-affiliated hospitals. PATIENTS Brain-dead adult patients managed in the participating ICUs over a 4-year period. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Among the 231 included patients, 145 (62.8%) were classified as ideal or extended-criteria potential lung donors at ICU admission and the remaining 86 patients having baseline contraindication for donation. Culture-proven aspiration pneumonia and early-onset ventilator-associated pneumonia occurred in 54 patients (23.4%) and 15 patients (6.5%), respectively (overall pneumonia incidence, 29.9%). Staphylococcus aureus and Enterobacterales were the most common pathogens. Using mixed-effects Cox proportional hazard models, age (adjusted hazard ratio, 0.98; 95% CI [0.96-0.99]), anoxic brain injury (3.55 [1.2-10.5]), aspiration (2.29 [1.22-4.29]), and not receiving antimicrobial agents at day 1 (3.56 [1.94-6.53]) were identified as independent predictors of pneumonia occurrence in the whole study population. Analyses restricted to potential lung donors yielded similar results. Pneumonia was associated with a postadmission decrease in the PaO2/FIO2 ratio and lower values at brain death, in the whole study population (estimated marginal mean, 294 [264-323] vs 365 [346-385] mm Hg in uninfected patients; p = 0.0005) as in potential lung donors (299 [248-350] vs 379 [350-408] mm Hg; p = 0.04; linear mixed models). Lungs were eventually retrieved in 31 patients (34.4%) among the 90 potential lung donors with at least one other organ harvested (pneumonia prevalence in lung donors (9.7%) vs nondonors (49.2%); p = 0.0002). CONCLUSIONS Pneumonia occurs in one-third of brain-dead patients and appears as the main reason for lung nonharvesting in those presenting as potential lung donors. The initiation of antimicrobial prophylaxis upon the first day of the ICU stay in comatose patients with severe brain injury could enlarge the pool of actual lung donors.
Collapse
|
19
|
Wei M, Huang Q, Liu Z, Luo Y, Xia J. Intestinal Barrier Dysfunction Participates in the Pathophysiology of Ischemic Stroke. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:401-416. [PMID: 33749565 DOI: 10.2174/1871527320666210322115808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/12/2020] [Accepted: 11/30/2020] [Indexed: 11/22/2022]
Abstract
The gastrointestinal tract is a major organ for the body to absorb nutrients, water and electrolytes. At the same time, it is a tight barrier to resist the invasion of harmful substances and maintain the homeostasis of the internal environment. Destruction of the intestinal barrier is linked to the digestive system, cardiovascular system, endocrine system and other systemic diseases. Mounting evidence suggests that ischemic stroke not only changes the intestinal microbes, but also increases the permeability of the intestinal barrier, leading to bacterial translocation, infection, and even sepsis. The intestinal barrier, as part of the gut-brain axis, has also been proven to participate in the pathophysiological process of ischemic stroke. However, little attention has been paid to it. Since ischemic stroke is a major public health issue worldwide, there is an urgent need to know more about the disease for better prevention, treatment and prognosis. Therefore, understanding the pathophysiological relationship between ischemic stroke and the intestinal barrier will help researchers further uncover the pathophysiological mechanism of ischemic stroke and provide a novel therapeutic target for the treatment of ischemic stroke. Here, we review the physiology and pathology between ischemic stroke and intestinal barrier based on related articles published in the past ten years about the relationship between ischemic stroke, stroke risk factors and intestinal flora, intestinal barrier, and discuss the following parts: the intestinal barrier; possible mechanisms of intestinal barrier destruction in ischemic stroke; intestinal barrier destruction caused by stroke-related risk factors; intestinal barrier dysfunction in ischemic stroke; targeting the intestinal barrier to improve stroke; conclusions and perspectives.
Collapse
Affiliation(s)
- Minping Wei
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Qin Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Zeyu Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Yunfang Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| |
Collapse
|
20
|
Xing D, Chen YH, Wang LT, Yu B, Ran ZB, Chen L. Evaluation of the therapeutic effect of high-flow nasal cannula oxygen therapy on patients with aspiration pneumonia accompanied by respiratory failure in the post-stroke sequelae stage. BMC Pulm Med 2021; 21:17. [PMID: 33413281 PMCID: PMC7788538 DOI: 10.1186/s12890-020-01359-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022] Open
Abstract
Background The aim of the present study was to evaluate the therapeutic effect of high-flow nasal cannula (HFNC) oxygen therapy on patients with aspiration pneumonia accompanied by respiratory failure in the post-stroke sequelae stage, with the goal of providing more effective oxygen therapy and improving patient prognosis. Methods Retrospective analysis was conducted on 103 elderly patients with post-stroke aspiration pneumonia and moderate respiratory failure (oxygenation index: 100–200 mmHg) that had been admitted. The patients were divided into two groups according to the mode of oxygen therapy that was used: the Venturi mask group and the HFNC treatment group. The two groups were analyzed and compared in terms of the changes in the blood gas indices measured at different points in time (4, 8, 12, 24, 48, and 72 h), the proportion of patients that required transition to invasive auxiliary ventilation, and the 28-day mortality rate. Results A total of 103 patients were retrospectively analyzed; 16 cases were excluded, and 87 patients were included in the final patient group (42 in the HFNC group and 45 in the Venturi group). There was a statistically significant difference in the oxygenation indices of the HFNC group and the Venturi group (F = 546.811, P < 0.05). There was a statistically significant interaction between the monitored oxygenation indices and the mode of oxygen therapy (F = 70.961, P < 0.05), and there was a statistically significant difference in the oxygenation indices for the two modes of oxygen therapy (F = 256.977, P < 0.05). HFNC therapy contributed to the improvement of the oxygenation indices at a rate of 75.1%. The Venturi and HFNC groups also differed significantly in terms of the proportion of patients that required transition to invasive auxiliary ventilation within 72 h (P < 0.05). The HFNC group’s risk for invasive ventilation was 0.406 times that of the Venturi group (P < 0.05). There was no statistical difference in the 28-day mortality rate of the two groups (P > 0.05). Conclusion HFNC could significantly improve the oxygenation state of patients with post-stroke aspiration pneumonia and respiratory failure, and it may reduce the incidence of invasive ventilation.
Collapse
Affiliation(s)
- Dong Xing
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yu-Hong Chen
- Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lan -Tao Wang
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Bin Yu
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Zhi -Bin Ran
- Department of Neurology, Shijiazhuang Great Wall Hospital, Shijiazhuang, 050000, China
| | - Li Chen
- Department of General Medicine, The Fourth Hospital of Hebei Medical University, No. 12 of Jiankang Road, Chang'an District, Shijiazhuang, 050011, China.
| |
Collapse
|
21
|
Gradek-Kwinta E, Czyzycki M, Weglarczyk K, Slowik A, Siedlar M, Dziedzic T. Ex vivo synthesized cytokines as a biomarker of stroke-associated pneumonia. Clin Chim Acta 2020; 510:260-263. [DOI: 10.1016/j.cca.2020.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/28/2020] [Accepted: 07/17/2020] [Indexed: 10/23/2022]
|
22
|
Yu Z, Zhou H, Shan B, Fu J, Zhu H, Feng Q, Shen R, Jin X. Clopidogrel increases risk of pneumonia compared with aspirin in acute ischemic minor stroke patients. J Thromb Thrombolysis 2020; 52:301-307. [PMID: 33034814 DOI: 10.1007/s11239-020-02306-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 01/06/2023]
Abstract
Antiplatelet agents may increase the risk of infections via suppressing platelet-mediated immune response. Here we assessed the contribution of clopidogrel versus aspirin to the development of pneumonia during an acute ischemic stroke admission. A retrospective cohort study was conducted of acute ischemic stroke patients who were admitted to our hospital from 2015 to 2018. Included patients received uninterrupted clopidogrel or aspirin therapy and did not take other antiplatelet agents throughout their stay. The interest outcome was development of pneumonia after stroke. Conditional logistic regression model after propensity score matching and adjusted logistic regression model were used to assess the impact of clopidogrel versus aspirin on post-stroke pneumonia. Among 1470 included patients, 1135 received aspirin and 335 received clopidogrel. Total 149 patients (10.1%) experienced pneumonia during the stroke hospitalization period. No difference was observed between clopidogrel cohort and aspirin cohort in the incidence of post-stroke pneumonia after propensity score matching (relative risk, 1.04; 95% confidence interval (CI) 0.65-1.65; P = 0.875). However, we found that clopidogrel was associated with increased risk of pneumonia compared with aspirin in minor stroke patients (adjusted odds ratio, 2.21; 95% CI 1.12-4.34; P = 0.021), and a statistically insignificant increase of pneumonia in diabetics (adjusted odds ratio, 1.94; 95% CI 0.96-3.94; P = 0.065). Compared with aspirin, clopidogrel is associated with increased pneumonia in minor stroke patients among who the interference of stroke-induced immunosuppression is minimized. Hence, aspirin may be a better choice for minor stroke patients in acute phase of ischemic stroke when pneumonia most frequently occurs.
Collapse
Affiliation(s)
- Zhangfeng Yu
- Department of Emergency Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Hua Zhou
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Baoshuai Shan
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Jianzhong Fu
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Hao Zhu
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Qian Feng
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Rong Shen
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xing Jin
- Department of Clinical Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, 16 Baita West Road, Suzhou, 215001, Jiangsu Province, China.
| |
Collapse
|
23
|
Li XJ, You XY, Wang CY, Li XL, Sheng YY, Zhuang PW, Zhang YJ. Bidirectional Brain-gut-microbiota Axis in increased intestinal permeability induced by central nervous system injury. CNS Neurosci Ther 2020; 26:783-790. [PMID: 32472633 PMCID: PMC7366750 DOI: 10.1111/cns.13401] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/19/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Central nervous system injuries may lead to the disorders of the hypothalamic‐pituitary‐adrenal axis, autonomic nervous system, and enteric nervous system. These effects then cause the changes in the intestinal microenvironment, such as a disordered intestinal immune system as well as alterations of intestinal bacteria. Ultimately, this leads to an increase in intestinal permeability. Inflammatory factors produced by the interactions between intestinal neurons and immune cells as well as the secretions and metabolites of intestinal flora can then migrate through the intestinal barrier, which will aggravate any peripheral inflammation and the central nervous system injury. The brain‐gut‐microbiota axis is a complex system that plays a crucial role in the occurrence and development of central nervous system diseases. It may also increase the consequences of preventative treatment. In this context, here we have summarized the factors that can lead to the increased intestinal permeability and some of the possible outcomes.
Collapse
Affiliation(s)
- Xiao-Jin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin-Yu You
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Cong-Ying Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xue-Li Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuan-Yuan Sheng
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng-Wei Zhuang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin, China
| | - Yan-Jun Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin, China
| |
Collapse
|
24
|
Imbalance in the force: the dark side of the microbiota on stroke risk and progression. Curr Opin Neurobiol 2019; 62:10-16. [PMID: 31809996 DOI: 10.1016/j.conb.2019.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/26/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022]
Abstract
The composition of the gut microbiota depends on many factors, including our lifestyle, diet, metabolism, antibiotic use and hygiene. The contribution of these factors in shaping the gut microbiota and the subsequent effects on the prevention and development of stroke has been under intense investigation. Furthermore, several reports have uncovered the impact of stroke on intestinal dysfunction and gut dysbiosis, highlighting the delicate interplay between the brain, gut and microbiome following this acute brain injury. Despite our growing appreciation of the gut microbiota in shaping brain health, the immune system, host metabolism and disease progression, its therapeutic capability in stroke is yet to be fully exploited. This review will explore the microbiota-gut-brain axis in stroke, and examine the potential role of the gut microbiota in the onset, progression and recovery phase of stroke.
Collapse
|
25
|
Wen SW, Shim R, Ho L, Wanrooy BJ, Srikhanta YN, Prame Kumar K, Nicholls AJ, Shen SJ, Sepehrizadeh T, Veer M, Srikanth VK, Ma H, Phan TG, Lyras D, Wong CHY. Advanced age promotes colonic dysfunction and gut-derived lung infection after stroke. Aging Cell 2019; 18:e12980. [PMID: 31199577 PMCID: PMC6718525 DOI: 10.1111/acel.12980] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/16/2019] [Accepted: 05/12/2019] [Indexed: 12/15/2022] Open
Abstract
Bacterial infection a leading cause of death among patients with stroke, with elderly patients often presenting with more debilitating outcomes. The findings from our retrospective study, supported by previous clinical reports, showed that increasing age is an early predictor for developing fatal infectious complications after stroke. However, exactly how and why older individuals are more susceptible to infection after stroke remains unclear. Using a mouse model of transient ischaemic stroke, we demonstrate that older mice (>12 months) present with greater spontaneous bacterial lung infections compared to their younger counterparts (7–10 weeks) after stroke. Importantly, we provide evidence that older poststroke mice exhibited elevated intestinal inflammation and disruption in gut barriers critical in maintaining colonic integrity following stroke, including reduced expression of mucin and tight junction proteins. In addition, our data support the notion that the localized pro‐inflammatory microenvironment driven by increased tumour necrosis factor‐α production in the colon of older mice facilitates the translocation and dissemination of orally inoculated bacteria to the lung following stroke onset. Therefore, findings of this study demonstrate that exacerbated dysfunction of the intestinal barrier in advanced age promotes translocation of gut‐derived bacteria and contributes to the increased risk to poststroke bacterial infection.
Collapse
Affiliation(s)
- Shu Wen Wen
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Raymond Shim
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Luke Ho
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
- Department of Medicine (Academic Unit), Peninsula Clinical School, Central Clinical School Monash University Frankston Victoria Australia
| | - Brooke J. Wanrooy
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Yogitha N. Srikhanta
- Department of Microbiology, Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia
| | - Kathryn Prame Kumar
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Alyce J. Nicholls
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - SJ. Shen
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Tara Sepehrizadeh
- Monash Biomedical Imaging Monash University Clayton Victoria Australia
| | - Michael Veer
- Monash Biomedical Imaging Monash University Clayton Victoria Australia
| | - Velandai K. Srikanth
- Department of Medicine (Academic Unit), Peninsula Clinical School, Central Clinical School Monash University Frankston Victoria Australia
| | - Henry Ma
- Stroke and Ageing Research Group, Department of Medicine, School of Clinical Sciences, Monash Medical Centre Monash University Clayton Victoria Australia
| | - Thanh G. Phan
- Stroke and Ageing Research Group, Department of Medicine, School of Clinical Sciences, Monash Medical Centre Monash University Clayton Victoria Australia
| | - Dena Lyras
- Department of Microbiology, Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia
| | - Connie H. Y. Wong
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences Monash University Clayton Victoria Australia
| |
Collapse
|
26
|
Huang YY, Li X, Li X, Sheng YY, Zhuang PW, Zhang YJ. Neuroimmune crosstalk in central nervous system injury-induced infection and pharmacological intervention. Brain Res Bull 2019; 153:232-238. [PMID: 31536756 DOI: 10.1016/j.brainresbull.2019.09.003] [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: 06/30/2019] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 12/30/2022]
Abstract
Infection (such as pneumonia and urinary tract infection) is one of the leading causes of death in patients with acute central nervous system (CNS) injury, which also greatly affects the patients' prognosis and quality of life. Antibiotics are commonly used for the treatment of various infections, however, available evidence demonstrate that prophylactic antibiotic treatments for CNS injury-induced infection have been unsuccessful. Effective approaches for prevention of CNS injury induced-infection remain scarce, therefore, better understanding the molecular and cellular mechanisms of infection post-CNS injury may aid in the development of efficacious therapeutic options. CNS injury-induced infection is confirmed affected by the sympathetic/parasympathetic nervous system, hypothalamic-pituitary-adrenal axis, and even brain-gut axis. In this review, we summarized the mechanisms of CNS injury- induced infection, crosstalk between the CNS and the immune system and current pharmacological intervention to provide ideas for the development of new anti- infective therapeutic strategies.
Collapse
Affiliation(s)
- Ying-Ying Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Xueli Li
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Xiaojin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Yuan-Yuan Sheng
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Peng-Wei Zhuang
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| | - Yan-Jun Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, 301617, China.
| |
Collapse
|
27
|
Sharma R, Shultz SR, Robinson MJ, Belli A, Hibbs ML, O'Brien TJ, Semple BD. Infections after a traumatic brain injury: The complex interplay between the immune and neurological systems. Brain Behav Immun 2019; 79:63-74. [PMID: 31029794 DOI: 10.1016/j.bbi.2019.04.034] [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: 01/08/2019] [Revised: 03/29/2019] [Accepted: 04/24/2019] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious global health issue, being the leading cause of death and disability for individuals under the age of 45, and one of the largest causes of global neurological disability. In addition to the brain injury itself, it is increasingly appreciated that a TBI may also alter the systemic immune response in a way that renders TBI patients more vulnerable to infections in the acute post-injury period. Such infections pose an additional challenge to the patient, increasing rates of mortality and morbidity, and worsening neurological outcomes. Hospitalization, surgical interventions, and a state of immunosuppression induced by injury to the central nervous system (CNS), may all contribute to the high rate of infections seen in the population with TBI. Ongoing research to better understand the immunomodulators that underlie TBI-induced immunosuppression may aid in the development of effective therapeutic strategies to improve the recovery trajectory for patients. This review first describes the clinical scenario, posing the question of whether TBI patients are more susceptible to infections such as pneumonia, and if so, why? We then consider how cross-talk between the injured brain and the systemic immune system occurs, and further, how the additional immune challenge of an acquired infection can contribute to ongoing neuroinflammation and neurodegeneration after a TBI. Experimental models combining TBI with infection are discussed, as well as current treatment options available for this double-barreled insult. The aims of this review are to summarize current understanding of the bidirectional relationship between the CNS and the immune system when faced with a mechanical trauma combined with a concomitant infection, and to highlight key outstanding questions that remain in the field.
Collapse
Affiliation(s)
- Rishabh Sharma
- Department of Neuroscience, Central Clinical School at the Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School at the Alfred Hospital, Monash University, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
| | - Marcus J Robinson
- Department of Immunology and Pathology, Central Clinical School at the Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Antonio Belli
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School at the Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School at the Alfred Hospital, Monash University, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School at the Alfred Hospital, Monash University, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
28
|
Lechtenberg KJ, Meyer ST, Doyle JB, Peterson TC, Buckwalter MS. Augmented β2-adrenergic signaling dampens the neuroinflammatory response following ischemic stroke and increases stroke size. J Neuroinflammation 2019; 16:112. [PMID: 31138227 PMCID: PMC6540436 DOI: 10.1186/s12974-019-1506-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
Background Ischemic stroke provokes a neuroinflammatory response and simultaneously promotes release of epinephrine and norepinephrine by the sympathetic nervous system. This increased sympathetic outflow can act on β2-adrenergic receptors expressed by immune cells such as brain-resident microglia and monocyte-derived macrophages (MDMs), but the effect on post-stroke neuroinflammation is unknown. Thus, we investigated how changes in β2-adrenergic signaling after stroke onset influence the microglia/MDM stroke response, and the specific importance of microglia/MDM β2-adrenergic receptors to post-stroke neuroinflammation. Methods To investigate the effects of β2-adrenergic receptor manipulation on post-stroke neuroinflammation, we administered the β2-adrenergic receptor agonist clenbuterol to mice 3 h after the onset of photothrombotic stroke. We immunostained to quantify microglia/MDM numbers and proliferation and to assess morphology and activation 3 days later. We assessed stroke outcomes by measuring infarct volume and functional motor recovery and analyzed gene expression levels of neuroinflammatory molecules. Finally, we evaluated changes in cytokine expression and microglia/MDM response in brains of mice with selective knockout of the β2-adrenergic receptor from microglia and monocyte-lineage cells. Results We report that clenbuterol treatment after stroke onset causes enlarged microglia/MDMs and impairs their proliferation, resulting in reduced numbers of these cells in the peri-infarct cortex by 1.7-fold at 3 days after stroke. These changes in microglia/MDMs were associated with increased infarct volume in clenbuterol-treated animals. In mice that had the β2-adrenergic receptor specifically knocked out of microglia/MDMs, there was no change in morphology or numbers of these cells after stroke. However, knockdown of β2-adrenergic receptors in microglia and MDMs resulted in increased expression of TNFα and IL-10 in peri-infarct tissue, while stimulation of β2-adrenergic receptors with clenbuterol had the opposite effect, suppressing TNFα and IL-10 expression. Conclusions We identified β2-adrenergic receptor signaling as an important regulator of the neuroimmune response after ischemic stroke. Increased β2-adrenergic signaling after stroke onset generally suppressed the microglia/MDM response, reducing upregulation of both pro- and anti-inflammatory cytokines, and increasing stroke size. In contrast, diminished β2-adrenergic signaling in microglia/MDMs augmented both pro- and anti-inflammatory cytokine expression after stroke. The β2-adrenergic receptor may therefore present a therapeutic target for improving the post-stroke neuroinflammatory and repair process.
Collapse
Affiliation(s)
- Kendra J Lechtenberg
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Scott T Meyer
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Janelle B Doyle
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Todd C Peterson
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA. .,Department of Neurosurgery, Stanford School of Medicine, Palo Alto, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
Vogelgesang A, Witt C, Heuer C, Schulze J, Gellrich J, von Sarnowski B, Langner S, Dressel A, Ruhnau J. Clinical Improvement Following Stroke Promptly Reverses Post-stroke Cellular Immune Alterations. Front Neurol 2019; 10:414. [PMID: 31118917 PMCID: PMC6504832 DOI: 10.3389/fneur.2019.00414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/04/2019] [Indexed: 01/09/2023] Open
Abstract
Background and Purpose: Stroke induces immediate profound alterations of the peripheral immune system rendering patients more susceptible to post-stroke infections. The precise mechanisms maintaining stroke-induced immune alterations (SIIA) remain unknown. High-Mobility-Group-Protein B1 (HMGB-1) is elevated for at least 7 days post-stroke and has been suggested to mediate SIIA. Patients with rapid clinical recovery of neurological deficits rarely develop severe infections. We therefore investigated whether rapid neurological recovery (either spontaneous or secondary to neurovascular recanalization therapy) alters the course of SIIA. National Institutes of Health Stroke Scale (NIHSS) served as surrogate marker for neurological improvement. Methods: Fluorescence-activated cell sorting was used to define leukocyte subpopulations. C-reactive protein (CRP), procalcitonin (PCT), HMGB-1, GM-CSF; IFN-β, IFN-γ, IL-1β, IL-1RA, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, IL-17, IL-17F, IL-18, TNF-α, MIF, IL-8, MCP-1, MCP-4, MIP-3α, MIP-3β, Eotaxin, soluble IL-6 receptor, E-selectin, and P-selectin were analyzed by ELISA or Multiplex Assays. Serum miRNA expression changes were analyzed by qPCR. Results: Cellular parameters were similar in the improved and non-improved cohort on admission. In patients with rapid clinical recovery absolute and relative leukocyte, neutrophil, and lymphocyte numbers normalized promptly overnight. In contrast, HMGB-1 serum levels did not differ between the two groups. Nine miRNA were found to be differentially expressed between improved and non-improved patients. Conclusions: SIIA are detectable on admission of acute stroke patients. While it was assumed that post-stroke immunosuppression is rapidly reversed with improvement this is the first data set that shows that improvement actually is associated with a rapid reversal of SIIA demonstrating that SIIA require a constant signal to persist. The observation that HMGB-1 serum concentrations were similar in improved and non-improved cohorts argues against a role for this pro-inflammatory mediator in the maintenance of SIIA. Serum miRNA observed to be regulated in stroke in other publications was counter regulated with improvement in our cohort.
Collapse
Affiliation(s)
| | - Carl Witt
- Department of Neurology, University Medicine, Greifswald, Germany
| | - Christin Heuer
- Department of Neurology, University Medicine, Greifswald, Germany
| | - Juliane Schulze
- Department of Neurology, University Medicine, Greifswald, Germany
| | - Juliane Gellrich
- Department of Neurology, University Medicine, Greifswald, Germany
| | | | - Sönke Langner
- Department of Diagnostic Radiology and Neuroradiology, University Medicine, Greifswald, Germany
| | - Alexander Dressel
- Department of Neurology, University Medicine, Greifswald, Germany.,Department of Neurology, Carl-Thiem-Klinikum, Cottbus, Germany
| | - Johanna Ruhnau
- Department of Neurology, University Medicine, Greifswald, Germany
| |
Collapse
|