1
|
Lv R, Li F, Liu Y, Song M, Yuan J, Zhang G, Sun M, Zhang Y, Su X, Zhao Y, Dong J, Shi Y, Zhao L. Molecularly imprinted nanoparticles hitchhiking on neutrophils for precise treatment of ischemic stroke. J Colloid Interface Sci 2025; 689:137246. [PMID: 40056670 DOI: 10.1016/j.jcis.2025.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Ischemic stroke (IS), the most prevalent type of stroke worldwide, is associated with a variety of complex processes, including oxidative stress, apoptosis, and ferroptosis. Recent findings indicate that inhibiting iron overload as a key regulatory mechanism of ferroptosis profoundly influences the pathogenesis and treatment of IS. In addition, enhanced blood-brain barrier (BBB) penetration and precise targeting of the ischaemic site contribute to improved therapeutic outcomes in IS. In this study, we developed FeSO4 templated-molecularly imprinted nanoparticles (MINPs) with high-affinity recognition of ferrous ions (Fe2+). MINPs exhibited physicochemical properties that perfectly match the polarity and condensed structure of Fe2+, resulting in the effective and specific clearance of Fe2+ through efficient and selective adsorption both in vivo and in vitro. Moreover, MINPs hitchhiked circulating neutrophils, thereby facilitating their penetration through BBB and enhancing targeted delivery to the ischemic brain. Our results, supported by transcriptomic analysis, further elucidated the molecular mechanisms by which MINPs significantly inhibit ferroptosis while concurrently regulating apoptosis and inflammation, thereby conferring marked neuroprotection against IS.
Collapse
Affiliation(s)
- Ruizhen Lv
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Fang Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Yong Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Mingzhu Song
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Jiayu Yuan
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Ge Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Mengdi Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Yifei Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Xiangchen Su
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Yuting Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Jia Dong
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou, Liaoning, China; Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
2
|
Wang J, Gao S, Cui Y, Liu XZ, Chen XX, Hang CH, Li W. Remote Organ Damage Induced by Stroke: Molecular Mechanisms and Comprehensive Interventions. Antioxid Redox Signal 2025; 42:885-904. [PMID: 40170638 DOI: 10.1089/ars.2024.0720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Significance: Damage after stroke is not only limited to the brain but also often occurs in remote organs, including the heart, lung, liver, kidney, digestive tract, and spleen, which are frequently affected by complex pathophysiological changes. The organs in the human body are closely connected, and signals transmitted through various molecular substances could regulate the pathophysiological changes of remote organs. Recent Advances: The latest studies have shown that inflammatory response plays an important role in remote organ damage after stroke, and can aggravate remote organ damage by activating oxidative stress, sympathetic axis, and hypothalamic axis, and disturbing immunological homeostasis. Remote organ damage can also cause damage to the brain, aggravating inflammatory response and oxidative damage. Critical Issues: Therefore, an in-depth exploration of inflammatory and oxidative mechanisms and adopting corresponding comprehensive intervention strategies have become necessary to reduce damage to remote organs and promote brain protection. Future Directions: The comprehensive intervention strategy involves multifaceted treatment methods such as inflammation regulation, antioxidants, and neural stem cell differentiation. It provides a promising treatment alternative for the comprehensive recovery of stroke patients and an inspiration for future research and treatment. The various organs of the human body are interconnected at the molecular level. Only through comprehensive intervention at the molecular and organ levels can we save remote organ damage and protect the brain after stroke to the greatest extent. Antioxid. Redox Signal. 42, 885-904.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Sen Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yue Cui
- Neurosurgical Institute, Nanjing University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xun-Zhi Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Xiang-Xin Chen
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
3
|
Chokkalla AK, Arruri V, Mehta SL, Vemuganti R. Loss of Epitranscriptomic Modification N 6-Methyladenosine (m 6A) Reader YTHDF1 Exacerbates Ischemic Brain Injury in a Sexually Dimorphic Manner. Transl Stroke Res 2025; 16:831-847. [PMID: 38869772 PMCID: PMC12066170 DOI: 10.1007/s12975-024-01267-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
N6-Methyladenosine (m6A) is a neuronal-enriched, reversible post-transcriptional modification that regulates RNA metabolism. The m6A-modified RNAs recruit various m6A-binding proteins that act as readers. Differential m6A methylation patterns are implicated in ischemic brain damage, yet the precise role of m6A readers in propagating post-stroke m6A signaling remains unclear. We presently evaluated the functional significance of the brain-enriched m6A reader YTHDF1, in post-stroke pathophysiology. Focal cerebral ischemia significantly increased YTHDF1 mRNA and protein expression in adult mice of both sexes. YTHDF1-/- male, but not female, mice subjected to transient middle cerebral artery occlusion (MCAO) showed worsened motor function recovery and increased infarction compared to sex-matched YTHDF1+/+ mice. YTHDF1-/- male, but not female, mice subjected to transient MCAO also showed significantly perturbed expression of genes related to inflammation, and increased infiltration of peripheral immune cells into the peri-infarct cortex, compared with sex-matched YTHDF1+/+ mice. Thus, this study demonstrates a sexual dimorphism of YTHDF1 in regulating post-ischemic inflammation and pathophysiology. Hence, post-stroke epitranscriptomic regulation might be sex-dependent.
Collapse
Affiliation(s)
- Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
4
|
Chen L, Zhou H, Tang Z, Deng H, Li Z. Influential factors related to feeding disorders in preterm infants and the construction of predictive models. Front Pediatr 2025; 13:1562778. [PMID: 40438787 PMCID: PMC12116673 DOI: 10.3389/fped.2025.1562778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/22/2025] [Indexed: 06/01/2025] Open
Abstract
Objective To investigate the influencing factors associated with feeding disorders in preterm infants and to construct a prediction model. Methods 314 cases of preterm infants admitted to our hospital from January 2019 to December 2022 were retrospectively analyzed and divided into feeding disorder group and non-feeding disorder group according to the presence of feeding disorder at 37 weeks of corrected gestational age. Statistical analysis of children's general information, hospitalization measures, laboratory tests, feeding time, etc. Multifactorial Logistic regression analysis of the occurrence of feeding disorders related to the influence of factors, and the use of subjects to make a work characteristic curve to analyze the predictive value of the relevant factors on feeding disorders. Results Multifactorial logistic regression analysis suggested that lower birth gestational age, birth weight, white blood cell count, absolute value of monocytes, blood calcium value, Apgar score at 1 min after birth, and longer duration of noninvasive ventilation were risk factors for feeding disorders in preterm infants. ROC curve analysis suggested that the area under the curve of the feeding disorders was predicted by the combination of the above seven indexes to construct the feeding disorders prediction model The AUC was 0.866 (P < 0.001, 95% CI 0.801-0.932), and it had a maximum Yoden index of 0.699, an optimal cutoff value of 0.169, a sensitivity of 85.4%, a specificity of 84.5%, and a prediction accuracy of 91.4%. Conclusions Lower birth gestational age, birth weight, white blood cell count, absolute monocyte value, blood calcium value, low Apgar score at 1 min after birth, and prolonged noninvasive ventilation are risk factors for feeding disorders in preterm infants, and the present prediction model is a good predictor of the occurrence of feeding disorders in preterm infants.
Collapse
Affiliation(s)
- Lishan Chen
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou City, China
- Department of Rehabilitation Medicine, The First People’s Hospital of Foshan City, Foshan City, China
| | - Huichang Zhou
- Department of Rehabilitation Medicine, The First People’s Hospital of Foshan City, Foshan City, China
| | - Zhiming Tang
- Department of Rehabilitation Medicine, The First People’s Hospital of Foshan City, Foshan City, China
| | - Haiyin Deng
- Department of Rehabilitation Medicine, The First People’s Hospital of Foshan City, Foshan City, China
| | - Zhihao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou City, China
| |
Collapse
|
5
|
Liu H, Yang K, Wang S, Ge J. Advancements in research on the thrombo-inflammation mechanisms mediated by factor XII in ischemic stroke. J Thromb Thrombolysis 2025:10.1007/s11239-025-03101-6. [PMID: 40281266 DOI: 10.1007/s11239-025-03101-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
Ischemic stroke (IS) is a major cause of mortality and disability, with thrombo-inflammation constituting a core pathophysiological mechanism. This process is closely linked to coagulation cascade activation, endothelial injury, immune cell infiltration, and neuronal damage. Coagulation factor XII (FXII), a key mediator of the contact activation pathway, has emerged as a promising therapeutic target due to its dual role in pathological thrombosis and immune regulation, without compromising physiological hemostasis. However, the clinical translation of FXII-targeted therapies is hindered by paradoxical observations. Recent studies highlight that FXII's functional complexity stems from its structural and spatial heterogeneity: full-length FXII derived from the liver and short FXII mRNA isoforms expressed in neurons mediate distinct biological effects. While FXII contributes to neuroinflammation and vascular injury via endothelial-platelet-neutrophil interactions, neuron-derived FXII exhibits neuroprotective effects through HGF-mediated signaling pathways. Additionally, circulating FXIIa promotes vascular remodeling by enhancing endothelial growth factor (VEGF) release. This review summarizes the multifaceted regulatory mechanisms of FXII in IS, focusing on its structure, distribution, preclinical-clinical paradox, and current therapeutic strategies. Special emphasis is placed on its domain-specific functions and the neuroprotective effects of FXII.
Collapse
Affiliation(s)
- Han Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
- School of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
- Hunan Academy of Chinese Medicine, No. 142 Yuehua Roud, Changsha, Hunan, China
| | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China.
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, No. 300, Xueshi Road, Changsha, Hunan, China.
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China.
- Hunan Academy of Chinese Medicine, No. 142 Yuehua Roud, Changsha, Hunan, China.
| |
Collapse
|
6
|
Yamazaki R, Azuma M, Osanai Y, Kouki T, Inagaki T, Kakita A, Takao M, Ohno N. Type I collagen secreted in white matter lesions inhibits remyelination and functional recovery. Cell Death Dis 2025; 16:285. [PMID: 40221393 PMCID: PMC11993711 DOI: 10.1038/s41419-025-07633-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
White matter injury is caused by cerebral blood flow disturbances associated with stroke and demyelinating diseases such as multiple sclerosis. Remyelination is induced spontaneously after white matter injury, but progressive multiple sclerosis and white matter stroke are usually characterised by remyelination failure. However, the mechanisms underlying impaired remyelination in lesions caused by demyelination and stroke remain unclear. In the current study, we demonstrated that collagen fibres accumulated in the demyelinated lesions of multiple sclerosis patients (age range 23-80 years) and white matter lesions of stroke patients (age range 80-87 years), suggesting that the accumulation of collagen fibres correlates with remyelination failure in these lesions. To investigate the function of collagen fibres in the white matter lesions, we generated two types of white matter injury in mice. We induced focal demyelination by lysolecithin (LPC) injection and ischemic stroke by endothelin 1 (ET1) injection into the internal capsule. We found that type I collagen fibres were secreted in ET1-induced lesions with impaired white matter regeneration in the chronic phase of disease. We also showed that monocyte-derived macrophages that infiltrated into lesions from the peripheral blood produced type I collagen after white matter injury, and that type I collagen also exacerbated microglial activation, astrogliosis, and axonal injury. Finally, we demonstrated that oligodendrocyte differentiation and remyelination were inhibited in the presence of type I collagen after LPC-induced demyelination. These results suggest that type I collagen secreted by monocyte-derived macrophages inhibited white matter regeneration, and therefore, the modulation of type I collagen metabolism might be a novel therapeutic target for white matter injury.
Collapse
Affiliation(s)
- Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan.
| | - Morio Azuma
- Department of Pharmacology, Division of Molecular Pharmacology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yasuyuki Osanai
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tom Kouki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takeshi Inagaki
- Department of Anatomy, Division of Forensic Medicine, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Takao
- Department of Clinical Laboratory, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Tochigi, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| |
Collapse
|
7
|
Forszt D, Gerreth K, Karpienko K, Zalewska A, Hojan K, Marchewka R, Bielas M, Maciejczyk M. Salivary chemokines and growth factors in patients with ischemic stroke. Sci Rep 2025; 15:12676. [PMID: 40221607 PMCID: PMC11993640 DOI: 10.1038/s41598-025-97974-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 04/08/2025] [Indexed: 04/14/2025] Open
Abstract
Stroke is a serious health problem that affects an increasing number of people. As a result of the blockage of blood flow, tissue necrosis occurs in areas of the brain supplied by the damaged vessel, and leads to the development of inflammation. Changes that occur in the brain allow molecules to enter the blood, and it has been suggested that some can also penetrate the saliva. This study is the first to assess the profile of 25 chemokines and growth factors in the saliva of stroke survivors compared to a control group. 22 stroke survivors and 22 individuals matched by age and gender were enrolled in the study. Salivary chemokines and growth factors were assessed using the multiplex ELISA method. In the unstimulated saliva of stroke patients, we demonstrated significantly higher levels of chemotactic factors (CTACK/CCL27, IL-8/CXCL8, MIG/CXCL9, MIF) and growth factors (basic FGF, G-CSF, HGF, LIF, VEGF) compared to controls. The levels of MCP-3/CCL7, eotaxin/CCL11, IP-10/CXCL10, IL-3/MCGF, and PDGF-BB were lower in the saliva of the study group. The concentration of basic FGF negatively correlated with cognitive function as measured by the Addenbrooke's Cognitive Examination (ACE) scale (p = 0.007 r = - 0.56), while salivary IL-3 and LIF levels positively correlated with scores on the Functional Independence Measure (FIM) scale (p = 0.019 r = 0.53; p = 0.033 r = 0.47, respectively). Receiver Operating Characteristic (ROC) analysis showed that salivary basic FGF, HGF, IL-3 and LIF can distinguish ischemic stroke patients from the control group with high sensitivity and specificity. In conclusion, disruptions in chemokine and growth factor levels in saliva may suggest an inflammatory etiology of ischemic stroke. Salivary basic FGF, HGF, IL-3 and LIF could serve as potential biomarkers for stroke. Further research is needed to illuminate the differences in salivary inflammatory mediator profiles in stroke and to evaluate the diagnostic utility of chemokines and growth factors in clinical practice.
Collapse
Affiliation(s)
- Dominika Forszt
- Department of Risk Group Dentistry, Chair of Pediatric Dentistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Karolina Gerreth
- Department of Risk Group Dentistry, Chair of Pediatric Dentistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Kamila Karpienko
- Students Scientific Club "Biochemistry of Civilization Diseases" at the Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Bialystok, Poland
| | - Anna Zalewska
- Experimental Dentistry Laboratory, Department of Conservative Dentistry, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Hojan
- Department of Occupational Therapy, Poznan University of Medical Sciences, Poznan, Poland
- Department of Rehabilitation, Greater Poland Cancer Centre, Poznan, Poland
| | - Renata Marchewka
- Neurorehabilitation Ward, Greater Poland Provincial Hospital, 60-480, Poznan, Poland
| | - Marzena Bielas
- Department of Family Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Mickiewicza 2C Street, 15-089, Bialystok, Poland.
| |
Collapse
|
8
|
Li T, Kang X, Zhang S, Wang Y, He J, Li H, Shao C, Kang J. Integrating machine learning and multi-omics analysis to reveal nucleotide metabolism-related immune genes and their functional validation in ischemic stroke. Front Immunol 2025; 16:1561544. [PMID: 40207230 PMCID: PMC11979214 DOI: 10.3389/fimmu.2025.1561544] [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: 01/17/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Background Ischemic stroke (IS) is a major global cause of death and disability, linked to nucleotide metabolism imbalances. This study aimed to identify nucleotide metabolism-related genes associated with IS and explore their roles in disease mechanisms for new diagnostic and therapeutic strategies. Methods IS gene expression data were sourced from the GEO database. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were conducted in R, intersecting results with nucleotide metabolism-related genes. Functional enrichment and connectivity map (cMAP) analyses identified key genes and potential therapeutic agents. Core immune-related genes were determined using LASSO regression, SVM-RFE, and Random Forest algorithms. Immune cell infiltration levels and correlations were analyzed via CIBERSORT. Single-cell RNA sequencing (scRNA-seq) data and molecular docking assessed gene expression, localization, and gene-drug binding. In vivo experiments validated core gene expression. Results Thirty-three candidate genes were identified, mainly involved in immune and inflammatory responses. CFL1, HMCES, and GIMAP1 emerged as key immune-related genes, linked to immune cell infiltration and showing high diagnostic potential. cMAP analysis indicated these genes as drug targets. scRNA-seq clarified their expression and localization, and molecular docking confirmed strong drug binding. In vivo experiments validated their significant expression in IS. Conclusion This study underscores the role of nucleotide metabolism in IS, identifying CFL1, HMCES, and GIMAP1 as potential biomarkers and therapeutic targets, providing insights for IS diagnosis and therapy development.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongyan Li
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, Changchun, China
| | - Chen Shao
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, Changchun, China
| | - Jingsong Kang
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
9
|
Merza Mohammad TA, Al-Haideri M, Azeez Al-Naqshabandi A. Decoding the immune Response: Analyzing PBMCs in ischemic stroke and Evaluating the effects of Rivaroxaban on gene expression. Hum Immunol 2025; 86:111252. [PMID: 39903995 DOI: 10.1016/j.humimm.2025.111252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Ischemic stroke (IS) is primarily caused by intricate inflammatory pathways and is a major global reason for mortality and disability. Patients with atrial fibrillation are treated with Rivaroxaban, a direct factor Xa inhibitor, to avoid stroke. This study looks at how certain genes are expressed in individuals with IS and how Rivaroxaban affects these genes and proteins. METHODS Using gene expression data from the GEO database, dysregulated genes in IS patients were found. Peripheral blood mononuclear cells from 50 IS patients were used to measure the expression of CXCL8, CXCL2, and G0S2 90 days before and after Rivaroxaban therapy using RT-PCR and ELISA. The Enrichr online tool was used to perform a functional enrichment analysis. RESULTS GEO2R analysis revealed that CXCL8, CXCL2, and G0S2 were significantly upregulated in IS samples compared to controls. Following Rivaroxaban therapy, the mRNA and protein levels of these genes showed a marked reduction, indicating a potential anti-inflammatory effect. CONCLUSION Rivaroxaban may control inflammatory responses in patients with IS, according to the study, which also reveals important genes implicated in IS. These results demonstrate the possibility of focused treatment approaches to reduce inflammation brought on by stroke.
Collapse
Affiliation(s)
- Talar Ahmad Merza Mohammad
- College of pharmacy Hawler Medical University Kurdistan region Iraq; University of Kurdistan Hewlêr (UKH) School of Medicine/ pharmacy department Kurdistan Region-Erbil Iraq.
| | - Maysoon Al-Haideri
- University of Kurdistan Hewlêr (UKH) School of Medicine/ pharmacy department Kurdistan Region-Erbil Iraq
| | | |
Collapse
|
10
|
Jiang Y, Wang N, Liu J, Ren H, Jiang W, Lei Y, Fu X, Hao M, Lang X, Liu Y, Liu X, Li R, Li H. Intranasal delivery of hMSC-derived supernatant for treatment of ischemic stroke by inhibiting the pro-inflammatory polarization of neutrophils. Stem Cell Res Ther 2025; 16:43. [PMID: 39901221 PMCID: PMC11792558 DOI: 10.1186/s13287-025-04172-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Stem cells utilized for ischemic stroke treatment often display unstable homing capabilities and diminished activity in vivo, limiting their neuroprotective efficacy. Furthermore, the optimal delivery route for stem cells remains undetermined. While the cytokines secreted by stem cells show promise in modulating post-stroke inflammation, the direct application of these supernatants in ischemic stroke treatment and the underlying mechanisms are still unclear. METHODS Secretory supernatants (hMSC-L) and cell lysate products (hMSC-M) from primary human umbilical cord mesenchymal stem cells-cultured medium were administered intranasally to mice with cerebral ischemia. The neuroprotective effects of hMSC-L and hMSC-M were assessed with TTC staining, behavioral tests and pathological staining. Flow cytometry and qPCR evaluated the expression of immune cells and cytokines in the CNS and peripheral immune organs. In vitro, flow cytometry and ELISA measured the effects of hMSC-L and hMSC-M on N2 polarization and inflammatory cytokines expression in primary murine neutrophils. Western blot analysis determined the impact of hMSC-L and hMSC-M on the PPAR-γ/STAT6/SOCS1 pathway, which is crucial for N2 neutrophil polarization. RESULTS TTC staining, behavioral experiments, and pathological assessments reveal intranasal delivery of hMSC-L and hMSC-M significantly reduces the infarct volume of mice with cerebral ischemia, improves neurological function scores, and promotes motor function recovery. Higher concentrations of hMSC-M contributed a more pronounced effect on neuropathological improvements in ischemic mice. Intranasal delivery of hMSC-L and hMSC-M significantly reduces neutrophil infiltration in the brain post-stroke and increases the proportion of anti-inflammatory N2-subtype neutrophils, boosting the expression levels of IL-10 and TGF-β. In vitro experiments demonstrate that hMSC-L and hMSC-M promote nuclear translocation of PPAR-γ in neutrophils stimulated with PMA, activating the downstream STAT6/SOCS1 signaling pathway to encourage N2-subtype neutrophil polarization. CONCLUSIONS Intranasal delivery of hMSC-L and hMSC-M effectively ameliorates cerebral ischemic injury in mice, comparable to traditional administration routes like intravenous delivery. Treatment with hMSC-L and hMSC-M enhances the PPAR-γ/STAT6/SOCS1 pathway and improves the neuroinflammatory response post-stroke by increasing N2 neutrophil infiltration. These results provide a theoretical basis for a deeper understanding of the mechanisms of stem cell therapy and for exploring suitable delivery pathways of stem cell treatment.
Collapse
Affiliation(s)
- Yixiang Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Ning Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Jingyi Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Haoran Ren
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Wenkang Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yanting Lei
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xidan Fu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Miao Hao
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xiujuan Lang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yumei Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xijun Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Rui Li
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350005, Fujian, China.
| | - Hulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
11
|
Chen X, Wang Y, Jin J, Jin P, Deng S. CCL17/CCR4 Axis Promotes Hematoma Clearance via ERK/AP1/SRA-Mediated Microglial Polarization After Intracerebral Hemorrhage. CNS Neurosci Ther 2025; 31:e70288. [PMID: 39996481 PMCID: PMC11851156 DOI: 10.1111/cns.70288] [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: 12/04/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Our previous studies demonstrated that CCL17 and its receptor CCR4 play crucial roles in neuroinflammation and microglial activation following intracerebral hemorrhage (ICH). However, the specific mechanisms by which the CCL17/CCR4 axis regulates microglial polarization and hematoma clearance remain unclear. AIMS This study investigates how the CCL17/CCR4 signaling pathway modulates microglial phenotype transition and enhances hematoma resolution after ICH, building upon our earlier findings showing CCR4's involvement in neuroinflammatory responses. METHODS Using CRISPR-mediated CCR4 disruption and CCR4 overexpression approaches in a mouse ICH model, we examined neurological outcomes, inflammatory responses, and hematoma volumes. We further evaluated the therapeutic potential of recombinant CCL17 administration. The downstream ERK signaling pathway's role in CCL17/CCR4-mediated microglial function was investigated through pharmacological inhibition. RESULTS CCR4 knockout exacerbated neurological deficits, increased neuroinflammation, and enlarged hematomas. In contrast, enhancing CCR4 expression or administering recombinant CCL17 improved functional recovery and provided neuroprotection. Mechanistically, CCL17/CCR4 signaling activated the ERK/AP1/SRA pathway, promoting anti-inflammatory, phagocytic microglial polarization, evidenced by increased CD206 and SRA expression. ERK inhibition reversed these protective effects. CONCLUSION Our findings extend previous work by revealing that the CCL17/CCR4 axis enhances hematoma clearance through the ERK/AP1/SRA pathway-mediated microglial polarization. This mechanism represents a promising therapeutic target for ICH treatment.
Collapse
Affiliation(s)
- Xianglan Chen
- Department of Critical Care MedicineHuaShan Hospital, Fudan UniversityShanghaiChina
| | - Yao Wang
- Department of Critical Care MedicineHuaShan Hospital, Fudan UniversityShanghaiChina
| | - Junjie Jin
- Department of Critical Care MedicineHuaShan Hospital, Fudan UniversityShanghaiChina
| | - Peng Jin
- Department of Intensive Care Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Shuixiang Deng
- Department of Critical Care MedicineHuaShan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
12
|
Wang C, Mao L, He M, Zhang J, Huang Y, Zhang Y, Xu J, Huang S, Gao Y. Caloric Restriction Preserves BBB Integrity After Transient Focal Cerebral Ischemia Through Reducing Neutrophil Infiltration. CNS Neurosci Ther 2025; 31:e70257. [PMID: 39915908 PMCID: PMC11802461 DOI: 10.1111/cns.70257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
AIMS Caloric restriction is a health-promoting lifestyle that has been reported to protect both white and gray matter in cases of ischemic stroke. This study will explore the underlying mechanism of restricted feeding (RF) and provide a theoretical basis for precise clinical treatment of stroke. METHODS In this study, we pretreated C57BL/6J mice with 70% RF for a continuous 28-day period prior to 60 min of transient focal cerebral ischemia (tFCI). Histological staining, diffusion tensor imaging (DTI), and behavioral assessments were used to assess RF's neuroprotection following tFCI. Immunofluorescence staining, quantitative real-time PCR, and flow cytometry were conducted to evaluate brain inflammation post-tFCI. Western blot, immunofluorescence staining, tracers, and electric microscopy were used to assess the blood-brain barrier (BBB) integrity. Peripheral neutrophils were cleared by administrating an anti-Ly6G antibody. RESULTS Initially, DTI, NeuN staining, and a batch of behavioral tests verified that RF significantly mitigated both gray/white matter injury and neurological deficits in the short- and long-term following tFCI. RF mice showed more anti-inflammatory microglia in their brains, along with reduced inflammatory cytokines, and chemokines. Interestingly, RF significantly reduced the neutrophils and macrophage infiltration. Subsequently, we observed that RF mice exhibited better BBB integrity following tFCI, with reduced neutrophil infiltration and matrix metalloprotein-9 release. Furthermore, the clearance of neutrophils with anti-Ly6G antibody in ad libitum feeding mice (LF-Ly6G) elicited comparable neuroprotective effects to those observed in RF, including improvements in neurological deficits, reductions in infarct volume, and mitigation of BBB damage. CONCLUSIONS In summary, our findings suggest that RF maintains the BBB integrity following ischemic stroke at least partially by reducing neutrophil infiltration, thereby alleviating both neurological and histological impairments.
Collapse
Affiliation(s)
- Chenran Wang
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Leilei Mao
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Miao He
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Jia Zhang
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yichen Huang
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yue Zhang
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Jing Xu
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Shaoqiang Huang
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yanqin Gao
- Department of Anesthesiology of Eye & Ent Hospital, Department of Anesthesiology of Obstetrics & Gynecology Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
13
|
Zheng Y, Guo Z, Wang J, Wu Z, Chen X, Zhu Y, Shan G, Hou H, Li X. Exploring and validating associations between six systemic inflammatory indices and ischemic stroke in a middle-aged and old Chinese population. Aging Clin Exp Res 2025; 37:31. [PMID: 39838216 PMCID: PMC11750927 DOI: 10.1007/s40520-024-02912-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Inflammation and maladaptive immune mechanisms have been substantiated as integral components in the critical pathological processes of the injury cascade in ischemic stroke (IS). This study aimed to explore the associations between six systemic inflammatory indices and IS in a Chinese population. METHODS This was a case-control study based on the retrospective review of electronic medical records from two hospitals in Shandong Province, China. Systemic inflammatory indices, including the systemic inflammation response index (SIRI), systemic immune inflammation index (SII), pan-immune-inflammation value (PIV), neutrophil lymphocyte ratio (NLR), platelet lymphocyte ratio (PLR), and lymphocyte monocyte ratio (LMR), were calculated. Logistic regression models and classification analyses were employed to evaluate associations and discriminatory abilities. RESULTS In total, 9392 participants aged 40-83 years old were included in the discovery (3620 pairs of IS-present cases and healthy controls) and validation (1076 pairs of IS-present cases and IS-absent controls with IS mimics) datasets. After adjusting for potential confounding factors, IS was found to be associated with all six systemic indices in the discovery dataset, including SIRI (odd ratio [OR] 8.77, 95% confidence interval [CI] 7.48-10.33), SII (1.03, 1.01-1.04), PIV (1.01, 1.01-1.01), NLR (2.23, 2.08-2.39), PLR (1.01, 1.01-1.01), and LMR (0.77, 0.75-0.78). Notably, only LMR exhibited significant associations with IS in both discovery and validation datasets (0.88, 0.83-0.93), suggesting an independent protective role of this index. SIRI, SII, PIV, NLR, and LMR showed good discriminative ability between IS patients and healthy controls in the discovery dataset (AUCs > 0.70). However, they performed poorly in distinguishing IS patients from IS mimics in the validation dataset (AUCs < 0.60). CONCLUSION This study provides valuable insights into the associations between systemic inflammatory indices and IS, offering potential implications for risk stratification. While these inflammatory indices are potential indicators for distinguishing IS from healthy conditions, additional biomarkers may be needed when differentiating IS from other chronic inflammatory conditions in clinical practice.
Collapse
Affiliation(s)
- Yulu Zheng
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
| | - Zheng Guo
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
| | | | - Zhiyuan Wu
- Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Xiaolin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yahong Zhu
- School of Science, Edith Cowan University, Perth, WA, Australia
| | - Guangle Shan
- Department of Bioinformatics, Thrive Bioresearch, Beijing, China
| | - Haifeng Hou
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia.
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong, China.
- The Second Affiliation Hospital of Shandong First Medical University, Tai'an, Shandong, China.
| | - Xingang Li
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia.
| |
Collapse
|
14
|
Yu T, Jiang P. Exploring the Role of Chemokine-Related Gene Deregulation and Immune Infiltration in Ischemic Stroke: Insights into CXCL16 and SEMA3E as Potential Biomarkers. J Mol Neurosci 2024; 74:115. [PMID: 39663269 DOI: 10.1007/s12031-024-02295-3] [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: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Ischemic stroke is a leading cause of mortality and disability globally. Understanding the role of chemokine-related differently expressed genes (CDGs) in ischemic stroke pathophysiology is essential for advancing diagnostic and therapeutic strategies. We conducted comprehensive analyses using the GSE16561 dataset: chemokine pathway enrichment via GSVA, differential expression of 12 CDGs, Pearson correlation, and functional enrichment analyses (GO and KEGG). Machine learning algorithms were employed to develop diagnostic models, evaluated using ROC curve analysis. A nomogram was constructed and validated with independent datasets (GSE58294). Gene set enrichment analysis (GSEA) and immuno-infiltration analysis were also performed. Chemokine pathway scores were significantly elevated in ischemic stroke, indicating their potential involvement. Logistic regression emerged as the most effective diagnostic model, with CXCL16 and SEMA3E as significant biomarkers. The nomogram exhibited high discriminatory ability (AUC = 0.964), well-calibrated predictions, and clinical utility across datasets. GSEA highlighted key biological pathways associated with CXCL16 and SEMA3E. Immuno-infiltration analysis revealed significant differences in immune cell infiltration between control and ischemic stroke groups, with distinct correlations between CXCL16 and SEMA3E expression and immune cell populations. This study highlights the deregulation of CDGs in ischemic stroke and their implications in critical biological processes. CXCL16 and SEMA3E are identified as key biomarkers with potential diagnostic utility. Insights from gene set enrichment and immuno-infiltration analyses provide mechanistic understanding, suggesting novel therapeutic targets and enhancing clinical decision-making in ischemic stroke management.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Anaesthesiology, Shandong Provincial Third Hospital, Shandong University, Tianqiao District, No. 12, Middle Wuyingshan Road, Jinan, Shandong Province, China
| | - Peng Jiang
- Department of Anaesthesiology, Shandong Provincial Third Hospital, Shandong University, Tianqiao District, No. 12, Middle Wuyingshan Road, Jinan, Shandong Province, China.
| |
Collapse
|
15
|
Carter K, Shah E, Waite J, Rana D, Zhao ZQ. Pathophysiology of Angiotensin II-Mediated Hypertension, Cardiac Hypertrophy, and Failure: A Perspective from Macrophages. Cells 2024; 13:2001. [PMID: 39682749 PMCID: PMC11640308 DOI: 10.3390/cells13232001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Heart failure is a complex syndrome characterized by cardiac hypertrophy, fibrosis, and diastolic/systolic dysfunction. These changes share many pathological features with significant inflammatory responses in the myocardium. Among the various regulatory systems that impact on these heterogeneous pathological processes, angiotensin II (Ang II)-activated macrophages play a pivotal role in the induction of subcellular defects and cardiac adverse remodeling during the progression of heart failure. Ang II stimulates macrophages via its AT1 receptor to release oxygen-free radicals, cytokines, chemokines, and other inflammatory mediators in the myocardium, and upregulates the expression of integrin adhesion molecules on both monocytes and endothelial cells, leading to monocyte-endothelial cell-cell interactions. The transendothelial migration of monocyte-derived macrophages exerts significant biological effects on the proliferation of fibroblasts, deposition of extracellular matrix proteins, induction of perivascular/interstitial fibrosis, and development of hypertension, cardiac hypertrophy and heart failure. Inhibition of macrophage activation using Ang II AT1 receptor antagonist or depletion of macrophages from the peripheral circulation has shown significant inhibitory effects on Ang II-induced vascular and myocardial injury. The purpose of this review is to discuss the current understanding in Ang II-induced maladaptive cardiac remodeling and dysfunction, particularly focusing on molecular signaling pathways involved in macrophages-mediated hypertension, cardiac hypertrophy, fibrosis, and failure. In addition, the challenges remained in translating these findings to the treatment of heart failure patients are also addressed.
Collapse
Affiliation(s)
| | | | | | | | - Zhi-Qing Zhao
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA 31404, USA
| |
Collapse
|
16
|
Wang H, Yang J, Cai Y, Zhao Y. Macrophages suppress cardiac reprogramming of fibroblasts in vivo via IFN-mediated intercellular self-stimulating circuit. Protein Cell 2024; 15:906-929. [PMID: 38530808 PMCID: PMC11637486 DOI: 10.1093/procel/pwae013] [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: 11/29/2023] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Direct conversion of cardiac fibroblasts (CFs) to cardiomyocytes (CMs) in vivo to regenerate heart tissue is an attractive approach. After myocardial infarction (MI), heart repair proceeds with an inflammation stage initiated by monocytes infiltration of the infarct zone establishing an immune microenvironment. However, whether and how the MI microenvironment influences the reprogramming of CFs remains unclear. Here, we found that in comparison with cardiac fibroblasts (CFs) cultured in vitro, CFs that transplanted into infarct region of MI mouse models resisted to cardiac reprogramming. RNA-seq analysis revealed upregulation of interferon (IFN) response genes in transplanted CFs, and subsequent inhibition of the IFN receptors increased reprogramming efficiency in vivo. Macrophage-secreted IFN-β was identified as the dominant upstream signaling factor after MI. CFs treated with macrophage-conditioned medium containing IFN-β displayed reduced reprogramming efficiency, while macrophage depletion or blocking the IFN signaling pathway after MI increased reprogramming efficiency in vivo. Co-IP, BiFC and Cut-tag assays showed that phosphorylated STAT1 downstream of IFN signaling in CFs could interact with the reprogramming factor GATA4 and inhibit the GATA4 chromatin occupancy in cardiac genes. Furthermore, upregulation of IFN-IFNAR-p-STAT1 signaling could stimulate CFs secretion of CCL2/7/12 chemokines, subsequently recruiting IFN-β-secreting macrophages. Together, these immune cells further activate STAT1 phosphorylation, enhancing CCL2/7/12 secretion and immune cell recruitment, ultimately forming a self-reinforcing positive feedback loop between CFs and macrophages via IFN-IFNAR-p-STAT1 that inhibits cardiac reprogramming in vivo. Cumulatively, our findings uncover an intercellular self-stimulating inflammatory circuit as a microenvironmental molecular barrier of in situ cardiac reprogramming that needs to be overcome for regenerative medicine applications.
Collapse
Affiliation(s)
- Hao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junbo Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yihong Cai
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
17
|
Hu X, Huang X, Yin T, Chen J, Zhao W, Yu M, Liu L, Du M. CX3CL1 (Fractalkine): An important cytokine in physiological and pathological pregnancies. J Reprod Immunol 2024; 166:104392. [PMID: 39577056 DOI: 10.1016/j.jri.2024.104392] [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: 09/09/2024] [Revised: 10/25/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024]
Abstract
C-X3-C motif chemokine ligand 1 (CX3CL1), commonly known as Fractalkine, is an important chemokine with dual functions of chemotaxis and adhesion. It plays a pivotal role in a variety of physiological processes and pathological conditions, particularly in conjunction with its receptor, C-X3-C motif chemokine receptor 1 (CX3CR1). This review focuses on the expression and intricate regulatory mechanisms of CX3CL1 at the maternal-fetal interface, emphasizing its multifaceted role during pregnancy. CX3CL1 was detected in the trophoblast and decidua tissues, playing a crucial role in recruitment of immune cells, enhancing endometrial receptivity, and modulating trophoblast cell activities. Abnormal expression of CX3CL1 has been correlated with adverse pregnancy outcomes such as spontaneous abortion, gestational diabetes, preeclampsia, and preterm births. By elucidating the complex interplay of CX3CL1 at the maternal-fetal interface, this review aims to shed light on its potential roles in pregnancy-related complications.
Collapse
Affiliation(s)
- Xianyang Hu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Xixi Huang
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Tingxuan Yin
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Jiajia Chen
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Weijie Zhao
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Min Yu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China.
| | - Lu Liu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China.
| | - Meirong Du
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China; Department of Obstetrics and Gynecology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
18
|
Chang YZ, Song YQ, Zhu HY, Zhang JR, Fu XG, Wang YL, Dong KH, Jiang CH, Mo DP, Zhang YP. Enhanced T-cell activation and chemokine-associated function in CD14-positive cells from venous sinus blood in sub-acute cerebral venous sinus thrombosis. Front Cell Dev Biol 2024; 12:1488005. [PMID: 39605979 PMCID: PMC11599252 DOI: 10.3389/fcell.2024.1488005] [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: 08/30/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Background Patients with sub-acute cerebral venous sinus thrombosis experience (SA.CVST) severe symptoms compared to two other venous sinus-related diseases, including chronic cerebral venous sinus thrombosis (C.CVST) and idiopathic intracranial hypertension (IIH). Objective This study aimed to determine whether the different immune reactions in different venous sinuses are related. Methods Stagnant blood in the cerebral venous sinuses was extracted by passing a microcatheter and CD14-positive cells were sorted by magnetic beads and subjected to RNA-seq sequencing. Results Compared to patients with IIH, 128 genes were significantly down-regulated and 373 genes were significantly up-regulated in the sub-acute CVST samples. The functions of these genes were mainly focused on "immune response", "T cell activation" and "plasma membrane". Gene Set Enrichment Analysis (GSEA) showed T cell survival and activation-related function significantly unregulated in sub-acute CVST. On the other hand, there were 366 genes down-regulated in chronic CVST and 75 genes up-regulated in chronic CVST. In functional annotation, these differently expressed genes were enriched in the "extracellular region", "chemokine-mediated signaling pathway" and "immune response". GSEA analysis confirmed that chemokine-related functions were all up-regulated in sub-acute CVST and monocyte-macrophage adhesion functions were also significantly up-regulated. Conclusion This study suggested the CD14-positive created an activated immune response in sub-acute CVST.
Collapse
Affiliation(s)
- Yu-Zhou Chang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu-Qi Song
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao-Yu Zhu
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jia-Rui Zhang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xi-Guang Fu
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yi-Long Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ke-Hui Dong
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chu-Han Jiang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Da-Peng Mo
- Interventional Neuroradiology Center, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu-Peng Zhang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
He Y, Wang J, Ying C, Xu KL, Luo J, Wang B, Gao J, Yin Z, Zhang Y. The interplay between ferroptosis and inflammation: therapeutic implications for cerebral ischemia-reperfusion. Front Immunol 2024; 15:1482386. [PMID: 39582857 PMCID: PMC11583640 DOI: 10.3389/fimmu.2024.1482386] [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: 08/18/2024] [Accepted: 10/14/2024] [Indexed: 11/26/2024] Open
Abstract
Stroke ranks as the second most significant contributor to mortality worldwide and is a major factor in disability. Ischemic strokes account for 71% of all stroke incidences globally. The foremost approach to treating ischemic stroke prioritizes quick reperfusion, involving methods such as intravenous thrombolysis and endovascular thrombectomy. These techniques can reduce disability but necessitate immediate intervention. After cerebral ischemia, inflammation rapidly arises in the vascular system, producing pro-inflammatory signals that activate immune cells, which in turn worsen neuronal injury. Following reperfusion, an overload of intracellular iron triggers the Fenton reaction, resulting in an excess of free radicals that cause lipid peroxidation and damage to cellular membranes, ultimately leading to ferroptosis. The relationship between inflammation and ferroptosis is increasingly recognized as vital in the process of cerebral ischemia-reperfusion (I/R). Inflammatory processes disturb iron balance and encourage lipid peroxidation (LPO) through neuroglial cells, while also reducing the activity of antioxidant systems, contributing to ferroptosis. Furthermore, the lipid peroxidation products generated during ferroptosis, along with damage-associated molecular patterns (DAMPs) released from ruptured cell membranes, can incite inflammation. Given the complex relationship between ferroptosis and inflammation, investigating their interaction in brain I/R is crucial for understanding disease development and creating innovative therapeutic options. Consequently, this article will provide a comprehensive introduction of the mechanisms linking ferroptosis and neuroinflammation, as well as evaluate potential treatment modalities, with the goal of presenting various insights for alleviating brain I/R injury and exploring new therapeutic avenues.
Collapse
Affiliation(s)
- Yuxuan He
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingyi Wang
- Faculty of Chinese Medicine of Macau University of Science and
Technology, Macao, Macao SAR, China
| | - Chunmiao Ying
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kang Li Xu
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingwen Luo
- Faculty of Chinese Medicine of Macau University of Science and
Technology, Macao, Macao SAR, China
| | - Baiqiao Wang
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Gao
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zaitian Yin
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yunke Zhang
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
21
|
Lu W, Wen J. Crosstalk Among Glial Cells in the Blood-Brain Barrier Injury After Ischemic Stroke. Mol Neurobiol 2024; 61:6161-6174. [PMID: 38279077 DOI: 10.1007/s12035-024-03939-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
Blood-brain barrier (BBB) is comprised of brain microvascular endothelial cells (ECs), astrocytes, perivascular microglia, pericytes, neuronal processes, and the basal lamina. As a complex and dynamic interface between the blood and the central nervous system (CNS), BBB is responsible for transporting nutrients essential for the normal metabolism of brain cells and hinders many toxic compounds entering into the CNS. The loss of BBB integrity following stroke induces tissue damage, inflammation, edema, and neural dysfunction. Thus, BBB disruption is an important pathophysiological process of acute ischemic stroke. Understanding the mechanism underlying BBB disruption can uncover more promising biological targets for developing treatments for ischemic stroke. Ischemic stroke-induced activation of microglia and astrocytes leads to increased production of inflammatory mediators, containing chemokines, cytokines, matrix metalloproteinases (MMPs), etc., which are important factors in the pathological process of BBB breakdown. In this review, we discussed the current knowledges about the vital and dual roles of astrocytes and microglia on the BBB breakdown during ischemic stroke. Specifically, we provided an updated overview of phenotypic transformation of microglia and astrocytes, as well as uncovered the crosstalk among astrocyte, microglia, and oligodendrocyte in the BBB disruption following ischemic stroke.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
22
|
Li J, Zhang L, Xue S, Yu C, Li Y, Li S, Ye Q, Duan X, Peng D. Exploration of the mechanism of Taohong Siwu Decoction for the treatment of ischemic stroke based on CCL2/CCR2 axis. Front Pharmacol 2024; 15:1428572. [PMID: 39268469 PMCID: PMC11390630 DOI: 10.3389/fphar.2024.1428572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Background and aims Taohong Siwu Decoction (THSWD) is a traditional Chinese herbal prescription that is effective for ischemic stroke, Whether THSWD regulates the CCL2/CCR2 axis and thus reduces the inflammatory response induced by ischemic stroke is not known. The aim of this study was to elucidate the mechanism of action of THSWD in the treatment of ischemic stroke using bioinformatics combined with in vitro and in vivo experiments. Methods R language was used to analyze middle cerebral artery occlusion/reperfusion (MCAO/R) rat transcriptome data and to identify differential gene expression following THSWD treatment. Gene set enrichment analysis (GSEA) was used to analyze the gene set enrichment pathway of MCAO/R rats treated with THSWD. PPI networks screened key targets. The Human Brain Microvascular Endothelial Cells (HBMEC) Oxygen Glucose Deprivation/Reoxygenation (OGD/R) model and SD rat models of MCAO/R were established. FITC-dextran, immunofluorescence, flow cytometry, ELISA, immunohistochemistry, Western blotting, and RT-qPCR were performed to identify potential treatment targets. Results A total of 515 differentially expressed genes of THSWD in MCAO/R rats were screened and 92 differentially expressed genes of THSWD potentially involved in stroke intervention were identified, including Cd68, Ccl2, and other key genes. In vitro, THSWD reversed the increase in permeability of HBMEC cells and M1/M2 polarization of macrophages induced by CCL2/CCR2 axis agonists. In vivo, THSWD improved nerve function injury and blood-brain barrier injury in MCAO/R rats. Further, THSWD inhibited the infiltration and polarization of macrophages, reduced the expression of IL-6, TNF-α, and MMP-9, and increased the expression of IL-4, while reducing the gene and protein expression of CCL2 and CCR2. Conclusion THSWD may play a protective role in ischemic stroke by inhibiting the CCL2/CCR2 axis, reducing the infiltration of macrophages, and promoting the polarization of M2 macrophages, thereby reducing inflammatory damage, and protecting injury to the blood-brain barrier.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lijuan Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Sujun Xue
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Chao Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yumeng Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Shuangping Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qingping Ye
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Key Laboratory of Chinese Medicinal Formula Research, Anhui University of Chinese Medicine, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Key Laboratory of Chinese Medicinal Formula Research, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
23
|
Liu L, Zhao B, Yu Y, Gao W, Liu W, Chen L, Xia Z, Cao Q. Vascular Aging in Ischemic Stroke. J Am Heart Assoc 2024; 13:e033341. [PMID: 39023057 PMCID: PMC11964078 DOI: 10.1161/jaha.123.033341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cellular senescence, a permanent halt in cell division due to stress, spurs functional and structural changes, contributing to vascular aging characterized by endothelial dysfunction and vascular remodeling. This process raises the risk of ischemic stroke (IS) in older individuals, with its mechanisms still not completely understood despite ongoing research efforts. In this review, we have analyzed the impact of vascular aging on increasing susceptibility and exacerbating the pathology of IS. We have emphasized the detrimental effects of endothelial dysfunction and vascular remodeling influenced by oxidative stress and inflammatory response on vascular aging and IS. Our goal is to aid the understanding of vascular aging and IS pathogenesis, particularly benefiting older adults with high risk of IS.
Collapse
Affiliation(s)
- Lian Liu
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Bo Zhao
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yueyang Yu
- Taikang Medical School, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Wenwei Gao
- Department of Critical Care MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| | - Weitu Liu
- Department of PathologyHubei Provincial Hospital of Traditional Chinese MedicineWuhanChina
| | - Lili Chen
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhongyuan Xia
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Quan Cao
- Department of NephrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
24
|
Shah FH, Lee HW. Endothelial and macrophage interactions in the angiogenic niche. Cytokine Growth Factor Rev 2024; 78:64-76. [PMID: 39019663 DOI: 10.1016/j.cytogfr.2024.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
The interactions between vascular cells, especially endothelial cells, and macrophages play a pivotal role in maintaining the subtle balance of vascular biology, which is crucial for angiogenesis in both healthy and diseased states. These cells are central to ensuring a harmonious balance between tissue repair and preventing excessive angiogenic activity, which could lead to pathological conditions. Recent advances in sophisticated genetic engineering vivo models and novel sequencing approaches, such as single-cell RNA-sequencing, in immunobiology have significantly enhanced our understanding of the gene expression and behavior of macrophages. These insights offer new perspectives on the role macrophages play not only in development but also across various health conditions. In this review, we explore the complex interactions between multiple types of macrophages and endothelium, focusing on their impact on new blood vessel formation. By understanding these intricate interactions, we aim to provide insights into new methods for managing angiogenesis in various diseases, thereby offering hope for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Fahad Hassan Shah
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Heon-Woo Lee
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea.
| |
Collapse
|
25
|
Ding R, Guan W, Yi M, Qin X, Wei S, Lu H, Wang Y, Lin C, Mei F, Xu H, Wu L. Identification of metabolic components of carotid plaque in high-risk patients utilizing liquid chromatography-tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9763. [PMID: 38745395 DOI: 10.1002/rcm.9763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 05/16/2024]
Abstract
OBJECTIVE Carotid atherosclerosis is a chronic progressive vascular disease that can be complicated by stroke in severe cases. Prompt diagnosis and treatment of high-risk patients are quite difficult due to the lack of reliable clinical biomarkers. This study aimed to explore potential plaque metabolic markers of stroke-prone risk and relevant targets for pharmacological intervention. METHOD Carotid intima and plaque sample tissues were obtained from 20 patients with cerebrovascular symptoms of carotid origin. An untargeted metabolomics approach based on liquid chromatography-tandem mass spectrometry was utilized to characterize the metabolic profiles of the tissues. Multivariate and univariate analysis tools were used. RESULTS A total of 154 metabolites were significantly altered in carotid plaque when compared with thickened intima. Of these, 62 metabolites were upregulated, whereas 92 metabolites were downregulated. Support vector machines identified the 15 most important metabolites, such as N-(cyclopropylmethyl)-N'-phenylurea, 9(S)-HOTrE, ACar 12:2, quinoxaline-2,3-dithiol, and l-thyroxine, as biomarkers for high-risk plaques. Metabolic pathway analysis showed that abnormal purine and nucleotide metabolism, amino acid metabolism, glutathione metabolism, and vitamin metabolism may contribute to the occurrence and progression of carotid atherosclerotic plaque. CONCLUSIONS Our study identifies the biomarkers and related metabolic mechanisms of carotid plaque, which is stroke-prone, and provides insights and ideas for the precise prevention and targeted intervention of the disease.
Collapse
Affiliation(s)
- Rui Ding
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenfei Guan
- Department of Vascular Surgery, Yichang Central People's Hospital, First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Man Yi
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohong Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shanshan Wei
- Department of Oncology, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Haoran Lu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuxuan Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chunnan Lin
- Department of Neurosurgery, Maoming People's Hospital, Maoming, Guangdong, China
| | - Fei Mei
- Department of Vascular Surgery, Yichang Central People's Hospital, First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Haitao Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Kucherova KS, Koroleva ES, Alifirova VM. The role of matrix metalloproteinases in the pathogenetic mechanisms of ischemic stroke. RUSSIAN NEUROLOGICAL JOURNAL 2024; 29:5-15. [DOI: 10.30629/2658-7947-2024-29-3-5-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Modern understanding of the mechanisms of the pathogenesis of ischemic stroke has expanded due to the study of neuroinfl ammation processes, in which matrix metalloproteinases (MMPs) play an important role. This literature review describes the main types of MMPs and provides current data on the pathophysiological role of this group of proteases in acute cerebral ischemia, which have multidirectional eff ects depending on the stage of the disease. Clinical studies assessing the role of MMPs in ischemic stroke are in most cases based on experimental models, and their results are ambiguous, which is determined by the versatility of their actions. MMPs are an important regulator of infl ammatory processes, the permeability of the blood-brain barrier and, as a consequence, cerebral edema. However, the positive eff ect of MMPs in the processes of angiogenesis, neurogenesis and neuroplasticity has been proven. Thus, further study of MMPs is relevant from the point of view of their role in functional recovery after ischemic stroke.
Collapse
|
27
|
Li X, Ma Y, Wang D. The role of P-selectin/PSGL-1 in regulating NETs as a novel mechanism in cerebral ischemic injury. Front Neurol 2024; 15:1442613. [PMID: 39022737 PMCID: PMC11252044 DOI: 10.3389/fneur.2024.1442613] [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: 06/02/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
In recent years, substantial advancements have been made in understanding the pathophysiology of ischemic stroke. Despite these developments, therapeutic options for cerebral ischemia remain limited due to stringent time windows and various contraindications. Consequently, there has been a concentrated effort to elucidate the underlying mechanisms of cerebral ischemic injury. Emerging research indicates that neutrophil extracellular traps (NETs) exacerbate inflammation and damage in ischemic brain tissue, contributing to neuronal cell death. The inhibition of NETs has shown potential in preventing thrombosis and the infiltration of immune cells. Central to the formation of NETs are P-selectin and its ligand, P-selectin glycoprotein ligand-1 (PSGL-1), which represent promising therapeutic targets. This review explores the detrimental impact of P-selectin, PSGL-1, and NETs on cerebral ischemia. Additionally, it delineates the processes by which P-selectin and PSGL-1 stimulate NETs production and provides evidence that blocking these molecules reduces NETs formation. This novel insight highlights a potential therapeutic avenue that warrants further investigation by researchers in the field.
Collapse
Affiliation(s)
- Xiao Li
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Yamin Ma
- Nanyang Hospital of Traditional Chinese Medicine, Nanyang, China
| | - Dongbin Wang
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, China
| |
Collapse
|
28
|
Zhao Z, Yan P, Zhang X, Yu X, Lv F, Gong M, Yang XA. Causal role of immune cells on cervical cancer onset revealed by two-sample Mendelian randomization study. Sci Rep 2024; 14:14890. [PMID: 38937531 PMCID: PMC11211447 DOI: 10.1038/s41598-024-65957-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024] Open
Abstract
Cervical cancer (CC) is a prevalent gynecological cancer worldwide that significantly impacts the quality of life and the physical and mental well-being of women. However, there have been limited studies utilizing Mendelian randomization (MR) analysis to investigate the connection between immune cells and CC. This study is to investigate the causal effects of immune traits on CC and non-neoplastic conditions of the cervix. The GWAS data for 731 immunophenotypes and six GWAS data for CC from the FinnGen database were downloaded. Subsequently, a two-sample MR analysis was conducted using the MR Egger, Weighted median, Inverse variance weighted (IVW), Simple mode, and Weighted mode methods. Our study has identified the potential causal effects of immune traits on inflammatory diseases of the cervix, other noninflammatory disorders of the cervix uteri, carcinoma in situ of cervix uteri, adenocarcinomas of cervix, squamous cell neoplasms and carcinoma of cervix, as well as malignant neoplasm of the cervix uteri, with the respective numbers being 8, 6, 11, 8, 23, and 12, respectively. A strong correlation between classic monocytes and various cervical diseases was revealed. Furthermore, we discovered that B cells expressing BAFF-R have the ability to impede the advancement of malignant CC, specifically squamous cell neoplasms and carcinoma of cervix. Our study has demonstrated a significant association between immune traits and both CC and non-neoplastic conditions of the cervix through two-sample Mendelian randomization, providing valuable insights for future clinical research.
Collapse
Affiliation(s)
- Zicheng Zhao
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China
| | - Pengxian Yan
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China
| | - Xiaoyu Zhang
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Xiaomin Yu
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China
| | - Fengchun Lv
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China
| | - Mingyu Gong
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China
| | - Xiu-An Yang
- Laboratory of Genetic Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, Hebei, China.
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, 067000, China.
| |
Collapse
|
29
|
Cao W, Song Y, Bai X, Yang B, Li L, Wang X, Wang Y, Chang W, Chen Y, Wang Y, Chen J, Gao P, Jiao L, Xu X. Systemic-inflammatory indices and clinical outcomes in patients with anterior circulation acute ischemic stroke undergoing successful endovascular thrombectomy. Heliyon 2024; 10:e31122. [PMID: 38778990 PMCID: PMC11109896 DOI: 10.1016/j.heliyon.2024.e31122] [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: 08/05/2023] [Revised: 04/01/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Background There is a lack of comprehensive profile assessment on complete blood count (CBC)-derived systemic-inflammatory indices, and their correlations with clinical outcome in patients with anterior circulation acute ischemic stroke (AIS) who achieved successful recanalization by endovascular thrombectomy (EVT). Methods Patients with anterior circulation AIS caused by large vessel occlusion (AIS-LVO) were retrospectively screened from December 2018 to December 2022. Systemic-inflammatory indices including ratios of neutrophil-to-lymphocyte (NLR), monocyte-to-lymphocyte (MLR), platelet-to-lymphocyte (PLR), and platelet-to-neutrophil (PNR), systemic immune-inflammation index (SII), systemic inflammation response index (SIRI), and aggregate inflammation systemic index (AISI) on admission and the first day post-EVT were calculated. Their correlations with symptomatic intracranial hemorrhage (sICH) and unfavorable 90-day functional outcome (modified Rankin Scale score of 3-6) were analyzed. Results A total of 482 patients [65 (IQR, 56-72) years; 33 % female] were enrolled, of which 231 (47.9 %) had unfavorable 90-day outcome and 50 (10.4 %) developed sICH. Day 1 neutrophil and monocyte counts, NLR, MLR, PLR, SII, SIRI, and AISI were increased, while lymphocyte and PNR were decreased compared to their admission levels. In multivariate analyses, neutrophil count, NLR, SII, and AISI on day 1 were independently associated with 90-day functional outcome. Moreover, day 1 neutrophil count, NLR, MLR, PLR, PNR, SII, and SIRI were independently linked to the occurrence of sICH. No admission variables were identified as independent risk factors for patient outcomes. Conclusion CBC-derived systemic-inflammatory indices measured on the first day after successful EVT are predictive of 90-day functional outcome and the sICH occurrence in patients with anterior circulation AIS-LVO.
Collapse
Affiliation(s)
- Wenbo Cao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
- Jinan Hospital of Xuanwu Hospital, Capital Medical University, 5106 Jingshi Road, Jinan, Shandong, 250100, China
| | - Yiming Song
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Bin Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
- Jinan Hospital of Xuanwu Hospital, Capital Medical University, 5106 Jingshi Road, Jinan, Shandong, 250100, China
| | - Xinyu Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Yuxin Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Wenxuan Chang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Yanfei Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Yabing Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Jian Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Peng Gao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
- Jinan Hospital of Xuanwu Hospital, Capital Medical University, 5106 Jingshi Road, Jinan, Shandong, 250100, China
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
- Jinan Hospital of Xuanwu Hospital, Capital Medical University, 5106 Jingshi Road, Jinan, Shandong, 250100, China
| |
Collapse
|
30
|
Bauer A, Boehme C, Mayer-Suess L, Rudzki D, Knoflach M, Kiechl S, Reindl M. Peripheral inflammatory response in people after acute ischaemic stroke and isolated spontaneous cervical artery dissection. Sci Rep 2024; 14:12063. [PMID: 38802464 PMCID: PMC11130263 DOI: 10.1038/s41598-024-62557-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/18/2024] [Indexed: 05/29/2024] Open
Abstract
The systemic inflammatory response following acute ischaemic stroke remains incompletely understood. We characterised the circulating inflammatory profile in 173 acute ischaemic stroke patients by measuring 65 cytokines and chemokines in plasma. Participants were grouped based on their inflammatory response, determined by high-sensitivity C-reactive protein levels in the acute phase. We compared stroke patients' profiles with 42 people experiencing spontaneous cervical artery dissection without stroke. Furthermore, variations in cytokine levels among stroke aetiologies were analysed. Follow-up samples were collected in a subgroup of ischaemic stroke patients at three and twelve months. Ischaemic stroke patients had elevated plasma levels of HGF and SDF-1α, and lower IL-4 levels, compared to spontaneous cervical artery dissection patients without stroke. Aetiology-subgroup analysis revealed reduced levels of nine cytokines/chemokines (HGF, SDF-1α, IL-2R, CD30, TNF-RII, IL-16, MIF, APRIL, SCF), and elevated levels of IL-4 and MIP-1β, in spontaneous cervical artery dissection (with or without ischaemic stroke as levels were comparable between both groups) compared to other aetiologies. The majority of cytokine/chemokine levels remained stable across the study period. Our research indicates that stroke due to large artery atherosclerosis, cardioembolism, and small vessel occlusion triggers a stronger inflammatory response than spontaneous cervical artery dissection.
Collapse
Affiliation(s)
- Angelika Bauer
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Boehme
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Mayer-Suess
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dagmar Rudzki
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Michael Knoflach
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Stefan Kiechl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
31
|
Ciechanowska A, Mika J. CC Chemokine Family Members' Modulation as a Novel Approach for Treating Central Nervous System and Peripheral Nervous System Injury-A Review of Clinical and Experimental Findings. Int J Mol Sci 2024; 25:3788. [PMID: 38612597 PMCID: PMC11011591 DOI: 10.3390/ijms25073788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Despite significant progress in modern medicine and pharmacology, damage to the nervous system with various etiologies still poses a challenge to doctors and scientists. Injuries lead to neuroimmunological changes in the central nervous system (CNS), which may result in both secondary damage and the development of tactile and thermal hypersensitivity. In our review, based on the analysis of many experimental and clinical studies, we indicate that the mechanisms occurring both at the level of the brain after direct damage and at the level of the spinal cord after peripheral nerve damage have a common immunological basis. This suggests that there are opportunities for similar pharmacological therapeutic interventions in the damage of various etiologies. Experimental data indicate that after CNS/PNS damage, the levels of 16 among the 28 CC-family chemokines, i.e., CCL1, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCL11, CCL12, CCL17, CCL19, CCL20, CCL21, and CCL22, increase in the brain and/or spinal cord and have strong proinflammatory and/or pronociceptive effects. According to the available literature data, further investigation is still needed for understanding the role of the remaining chemokines, especially six of them which were found in humans but not in mice/rats, i.e., CCL13, CCL14, CCL15, CCL16, CCL18, and CCL23. Over the past several years, the results of studies in which available pharmacological tools were used indicated that blocking individual receptors, e.g., CCR1 (J113863 and BX513), CCR2 (RS504393, CCX872, INCB3344, and AZ889), CCR3 (SB328437), CCR4 (C021 and AZD-2098), and CCR5 (maraviroc, AZD-5672, and TAK-220), has beneficial effects after damage to both the CNS and PNS. Recently, experimental data have proved that blockades exerted by double antagonists CCR1/3 (UCB 35625) and CCR2/5 (cenicriviroc) have very good anti-inflammatory and antinociceptive effects. In addition, both single (J113863, RS504393, SB328437, C021, and maraviroc) and dual (cenicriviroc) chemokine receptor antagonists enhanced the analgesic effect of opioid drugs. This review will display the evidence that a multidirectional strategy based on the modulation of neuronal-glial-immune interactions can significantly improve the health of patients after CNS and PNS damage by changing the activity of chemokines belonging to the CC family. Moreover, in the case of pain, the combined administration of such antagonists with opioid drugs could reduce therapeutic doses and minimize the risk of complications.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Str., 31-343 Kraków, Poland;
| |
Collapse
|
32
|
Saleh RO, Majeed AA, Margiana R, Alkadir OKA, Almalki SG, Ghildiyal P, Samusenkov V, Jabber NK, Mustafa YF, Elawady A. Therapeutic gene delivery by mesenchymal stem cell for brain ischemia damage: Focus on molecular mechanisms in ischemic stroke. Cell Biochem Funct 2024; 42:e3957. [PMID: 38468129 DOI: 10.1002/cbf.3957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024]
Abstract
Cerebral ischemic damage is prevalent and the second highest cause of death globally across patient populations; it is as a substantial reason of morbidity and mortality. Mesenchymal stromal cells (MSCs) have garnered significant interest as a potential treatment for cerebral ischemic damage, as shown in ischemic stroke, because of their potent intrinsic features, which include self-regeneration, immunomodulation, and multi-potency. Additionally, MSCs are easily obtained, isolated, and cultured. Despite this, there are a number of obstacles that hinder the effectiveness of MSC-based treatment, such as adverse microenvironmental conditions both in vivo and in vitro. To overcome these obstacles, the naïve MSC has undergone a number of modification processes to enhance its innate therapeutic qualities. Genetic modification and preconditioning modification (with medications, growth factors, and other substances) are the two main categories into which these modification techniques can be separated. This field has advanced significantly and is still attracting attention and innovation. We examine these cutting-edge methods for preserving and even improving the natural biological functions and therapeutic potential of MSCs in relation to adhesion, migration, homing to the target site, survival, and delayed premature senescence. We address the use of genetically altered MSC in stroke-induced damage. Future strategies for improving the therapeutic result and addressing the difficulties associated with MSC modification are also discussed.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Ali A Majeed
- Department of Pathological Analyses, Faculty of Science, University of Kufa, Najaf, Iraq
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ola Kamal A Alkadir
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Vadim Samusenkov
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Ahmed Elawady
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
33
|
Tynterova AM, Barantsevich ER, Shusharina NN. [Biomarkers of atherothrombotic and cardioembolic subtypes of acute ischemic stroke]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:20-26. [PMID: 39831358 DOI: 10.17116/jnevro202412412220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
OBJECTIVE To evaluate the concentrations of CC-chemokines and stable metabolites of nitric oxide (NO) and endothelin-1 (ET-1) in patients with atherothrombotic (AT) and cardioembolic (CE) subtypes of ischemic stroke (IS) in the acute period. MATERIAL AND METHODS Sixty patients diagnosed with IS in the carotid basin were examined. Group 1 included 30 patients with AT, group 2 - 30 patients with CE subtype of IS. The control group consisted of 20 age-matched patients without a history of stroke. All patients were assessed on admission for functional status using the Barthel Index (BI), the Modified Rankin Scale (mRS) and the National Institutes of Health Stroke Scale (NIHSS). Neuroimaging parameters were assessed using CT/MRI data. Laboratory diagnostics included assessment of serum concentrations of interleukin (IL)-1β, IL-6, IL-16, interferon gamma (IFN-γ), CC-chemokines (CCL2, CCL-7, CCL8, CCL13, CCL15, CCL23) and stable metabolites of NO and ET-1. RESULTS All patients with IS had moderate stroke severity scores according to NIHSS, BI and mRS. Analysis of the indices of the main clinical scales revealed. higher NIHSS scores, the prevalence of body mass index (BMI), the size of IS foci and the presence of multifocal lesions in patients of group 2. Compared with the control group, a significant increase of IFN- γ and IL-16 was noted in patients of both groups; ET-1, CCL2 and CCL15 were elevated in group 1; IL-1β, IL-6, CCL8 and CCL23 - in group 2. A comparative analysis between groups revealed higher concentrations of ET-1, CCL2 and CCL15 in group 1 and the increase of IL-1β, IL-6, IL-16 and CCL13 in group 2. CONCLUSION The results allow considering cytokines CCL23, IL-6, IL-16, IL-1β and IFN-γ as indicators of IS severity; CCL2, CCL15 and ET-1 as important regulators of atherogenesis and indicators of the AT subtype of IS; IL-1β, IL-6 and CCL13 as markers of complications of atrial fibrillation. The findings indicate the necessity of multicentre studies with a large sample size to determine the potential value of CCR1/CCR2 chemokines and stable metabolites as biomarkers of course of different IS subtypes.
Collapse
Affiliation(s)
- A M Tynterova
- Imannuel Kant Baltic Federal University, Kaliningrad, Russia
| | - E R Barantsevich
- Academian I.P. Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - N N Shusharina
- Imannuel Kant Baltic Federal University, Kaliningrad, Russia
| |
Collapse
|
34
|
Ronaldson PT, Williams EI, Betterton RD, Stanton JA, Nilles KL, Davis TP. CNS Drug Delivery in Stroke: Improving Therapeutic Translation From the Bench to the Bedside. Stroke 2024; 55:190-202. [PMID: 38134249 PMCID: PMC10752297 DOI: 10.1161/strokeaha.123.043764] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Drug development for ischemic stroke is challenging as evidenced by the paucity of therapeutics that have advanced beyond a phase III trial. There are many reasons for this lack of clinical translation including factors related to the experimental design of preclinical studies. Often overlooked in therapeutic development for ischemic stroke is the requirement of effective drug delivery to the brain, which is critical for neuroprotective efficacy of several small and large molecule drugs. Advancing central nervous system drug delivery technologies implies a need for detailed comprehension of the blood-brain barrier (BBB) and neurovascular unit. Such knowledge will permit the innate biology of the BBB/neurovascular unit to be leveraged for improved bench-to-bedside translation of novel stroke therapeutics. In this review, we will highlight key aspects of BBB/neurovascular unit pathophysiology and describe state-of-the-art approaches for optimization of central nervous system drug delivery (ie, passive diffusion, mechanical opening of the BBB, liposomes/nanoparticles, transcytosis, intranasal drug administration). Additionally, we will discuss how endogenous BBB transporters represent the next frontier of drug delivery strategies for stroke. Overall, this review will provide cutting edge perspective on how central nervous system drug delivery must be considered for the advancement of new stroke drugs toward human trials.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Erica I Williams
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Robert D Betterton
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Joshua A Stanton
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Kelsy L Nilles
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| |
Collapse
|
35
|
Yao Y, Liu F, Gu Z, Wang J, Xu L, Yu Y, Cai J, Ren R. Emerging diagnostic markers and therapeutic targets in post-stroke hemorrhagic transformation and brain edema. Front Mol Neurosci 2023; 16:1286351. [PMID: 38178909 PMCID: PMC10764516 DOI: 10.3389/fnmol.2023.1286351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/13/2023] [Indexed: 01/06/2024] Open
Abstract
Stroke is a devastating condition that can lead to significant morbidity and mortality. The aftermath of a stroke, particularly hemorrhagic transformation (HT) and brain edema, can significantly impact the prognosis of patients. Early detection and effective management of these complications are crucial for improving outcomes in stroke patients. This review highlights the emerging diagnostic markers and therapeutic targets including claudin, occludin, zonula occluden, s100β, albumin, MMP-9, MMP-2, MMP-12, IL-1β, TNF-α, IL-6, IFN-γ, TGF-β, IL-10, IL-4, IL-13, MCP-1/CCL2, CXCL2, CXCL8, CXCL12, CCL5, CX3CL1, ICAM-1, VCAM-1, P-selectin, E-selectin, PECAM-1/CD31, JAMs, HMGB1, vWF, VEGF, ROS, NAC, and AQP4. The clinical significance and implications of these biomarkers were also discussed.
Collapse
Affiliation(s)
- Ying Yao
- Department of Neuroscience Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fei Liu
- Department of Neuroscience Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhaowen Gu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lintao Xu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yue Yu
- Department of Neuroscience Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Cai
- Department of Neuroscience Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Reng Ren
- Department of Neuroscience Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
36
|
Marquez-Ortiz RA, Tesic V, Hernandez DR, Akhter B, Aich N, Boudreaux PM, Clemons GA, Wu CYC, Lin HW, Rodgers KM. Neuroimmune Support of Neuronal Regeneration and Neuroplasticity following Cerebral Ischemia in Juvenile Mice. Brain Sci 2023; 13:1337. [PMID: 37759938 PMCID: PMC10526826 DOI: 10.3390/brainsci13091337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Ischemic damage to the brain and loss of neurons contribute to functional disabilities in many stroke survivors. Recovery of neuroplasticity is critical to restoration of function and improved quality of life. Stroke and neurological deficits occur in both adults and children, and yet it is well documented that the developing brain has remarkable plasticity which promotes increased post-ischemic functional recovery compared with adults. However, the mechanisms underlying post-stroke recovery in the young brain have not been fully explored. We observed opposing responses to experimental cerebral ischemia in juvenile and adult mice, with substantial neural regeneration and enhanced neuroplasticity detected in the juvenile brain that was not found in adults. We demonstrate strikingly different stroke-induced neuroimmune responses that are deleterious in adults and protective in juveniles, supporting neural regeneration and plasticity. Understanding age-related differences in neuronal repair and regeneration, restoration of neural network function, and neuroimmune signaling in the stroke-injured brain may offer new insights for the development of novel therapeutic strategies for stroke rehabilitation.
Collapse
Affiliation(s)
- Ricaurte A. Marquez-Ortiz
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
| | - Vesna Tesic
- Department of Neurology, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA
| | - Daniel R. Hernandez
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
| | - Bilkis Akhter
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
| | - Nibedita Aich
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
| | - Porter M. Boudreaux
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
| | - Garrett A. Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA
| | - Hung Wen Lin
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
- Department of Neurology, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA
| | - Krista M. Rodgers
- Department of Cellular Biology and Anatomy, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA (B.A.)
- Department of Neurology, Louisiana State University, Health Sciences Center, Shreveport, LA 70803, USA
| |
Collapse
|