1
|
Rivera Antonio AM, Padilla Martínez II, Torres-Ramos MA, Rosales-Hernández MC. Myeloperoxidase as a therapeutic target for oxidative damage in Alzheimer's disease. J Enzyme Inhib Med Chem 2025; 40:2456282. [PMID: 39950933 DOI: 10.1080/14756366.2025.2456282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 05/09/2025] Open
Abstract
Alzheimer's disease (AD) is a major neurodegenerative disorder more common in older adults. One of the leading AD hypotheses involves the amyloid beta (A) production, it is associated to oxidative stress, neuroinflammation, and neurovascular damage. The interaction of A with the blood vessel wall contributes to the disruption of the blood-brain barrier (BBB), allowing neutrophil infiltration containing the myeloperoxidase enzyme (MPO), which produces hypochlorous acid (HOCl) a potent oxidant. Also, MPO could be released from the microglia cells and interact with the amyloid beta plaques. This review aims to study the role of MPO in the progression of AD, in particular its contribution to oxidative stress and neuroinflammation. Furthermore, to explore the MPO-potential as AD-biomarker to evaluate the therapeutic potential of its inhibitors to mitigate the neurotoxicity. Finally, revise MPO inhibitors that could act as dual inhibitors acting on MPO and acetylcholinesterase and or another target involved in AD.
Collapse
Affiliation(s)
- Astrid Mayleth Rivera Antonio
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, MéxicoCiudad de México, México
| | - Itzia Irene Padilla Martínez
- Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, MéxicoCiudad de México, México
| | - Mónica A Torres-Ramos
- Dirección de investigación del Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez. Av, Ciudad de México. C.P, México
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, MéxicoCiudad de México, México
| |
Collapse
|
2
|
Li J, Wang Z, Li J, Zhao H, Ma Q. HMGB1: A New Target for Ischemic Stroke and Hemorrhagic Transformation. Transl Stroke Res 2025; 16:990-1015. [PMID: 38740617 PMCID: PMC12045843 DOI: 10.1007/s12975-024-01258-5] [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: 02/29/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024]
Abstract
Stroke in China is distinguished by its high rates of morbidity, recurrence, disability, and mortality. The ultra-early administration of rtPA is essential for restoring perfusion in acute ischemic stroke, though it concurrently elevates the risk of hemorrhagic transformation. High-mobility group box 1 (HMGB1) emerges as a pivotal player in neuroinflammation after brain ischemia and ischemia-reperfusion. Released passively by necrotic cells and actively secreted, including direct secretion of HMGB1 into the extracellular space and packaging of HMGB1 into intracellular vesicles by immune cells, glial cells, platelets, and endothelial cells, HMGB1 represents a prototypical damage-associated molecular pattern (DAMP). It is intricately involved in the pathogenesis of atherosclerosis, thromboembolism, and detrimental inflammation during the early phases of ischemic stroke. Moreover, HMGB1 significantly contributes to neurovascular remodeling and functional recovery in later stages. Significantly, HMGB1 mediates hemorrhagic transformation by facilitating neuroinflammation, directly compromising the integrity of the blood-brain barrier, and enhancing MMP9 secretion through its interaction with rtPA. As a systemic inflammatory factor, HMGB1 is also implicated in post-stroke depression and an elevated risk of stroke-associated pneumonia. The role of HMGB1 extends to influencing the pathogenesis of ischemia by polarizing various subtypes of immune and glial cells. This includes mediating excitotoxicity due to excitatory amino acids, autophagy, MMP9 release, NET formation, and autocrine trophic pathways. Given its multifaceted role, HMGB1 is recognized as a crucial therapeutic target and prognostic marker for ischemic stroke and hemorrhagic transformation. In this review, we summarize the structure and redox properties, secretion and pathways, regulation of immune cell activity, the role of pathophysiological mechanisms in stroke, and hemorrhage transformation for HMGB1, which will pave the way for developing new neuroprotective drugs, reduction of post-stroke neuroinflammation, and expansion of thrombolysis time window.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Zixin Wang
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Jiameng Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Haiping Zhao
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| | - Qingfeng Ma
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| |
Collapse
|
3
|
Liu W, Wang YR, Wu H, Cui W, Xu X. The role of myeloperoxidase in the pathogenesis of stroke. Brain Res 2025; 1861:149705. [PMID: 40379076 DOI: 10.1016/j.brainres.2025.149705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/19/2025]
Abstract
Stroke is the leading cause of mortality and morbidity worldwide, significantly impacting human welfare and overall health. Myeloperoxidase (MPO), a heme peroxidase secreted by neutrophils, plays a crucial role in the body's defense mechanisms, exhibiting pro-inflammatory and pro-oxidative properties. Additionally, MPO compromises the structural integrity and functional capacity of blood vessels, potentially leading to the formation and dislodgement of atherosclerotic plaques, vascular stenosis, thrombosis, and ultimately contributing to stroke occurrence. Following a stroke, a significant influx of neutrophils infiltrates the cerebral tissue, leading to an excessive release of MPO-derived oxidants and the subsequent promotion of various inflammatory mediators, thereby exacerbating cerebral tissue damage. Numerous studies have consistently demonstrated the pivotal role of MPO in the pathogenesis and progression of stroke, establishing it as a reliable prognostic indicator. Exploring the association between MPO and stroke enhances our understanding of the pathological mechanisms underlying stroke and aids in the development of therapeutic interventions. This review provides a comprehensive analysis of the molecular structure and cellular localization of MPO, elucidating its critical role in mediating vascular injury, the formation of Neutrophil Extracellular Traps (NETs), oxidative stress, neuroinflammation, disruption of the blood-brain barrier (BBB), and neuronal apoptosis during stroke pathogenesis. Additionally, we discuss recent advancements in MPO-targeted drugs and Traditional Chinese Medicine compounds as potential therapeutic strategies for stroke treatment.
Collapse
Affiliation(s)
- Wei Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yi-Ran Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Hongyun Wu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| | - Wenqiang Cui
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| | - Xiangqing Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| |
Collapse
|
4
|
Espejo EF, Guerra MDM, Castellano S. Association between serum myeloperoxidase enzyme activity and Parkinson's disease status. NPJ Parkinsons Dis 2025; 11:94. [PMID: 40287421 PMCID: PMC12033337 DOI: 10.1038/s41531-025-00941-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Elevated levels of the inflammatory enzyme myeloperoxidase (MPO) in the blood are associated with the development of age-related inflammatory diseases. Given that age, inflammation, and blood MPO play a role in the pathogenesis of Parkinson´s disease (PD), we hypothesized that serum MPO could be associated with PD status. This case-control study (199 participants) was conducted using an extensive protocol, and the concentration and activity of MPO in blood serum were measured. The findings reveal that serum MPO concentration and activity are significantly increased in the patients, and that rates of PD in all individuals are associated with increasing tertiles of MPO concentration and activity. In multivariate logistic regression model adjusting for confounding factors, MPO activity (not concentration) is the factor that is most associated with PD status (OR, 6.921, P = 0.001). Mental depression is directly associated with MPO activity and with PD status (OR, 0.121, B = -2.108, P = 0.002). The use of statins or nonsteroidal anti-inflammatory drugs significantly reduces serum MPO activity, but the possible association with the odds of having PD does not survive correction for multiple testing. In summary, both serum MPO concentration and activity are increased in patients with PD, but only MPO enzyme activity is associated with PD status. These findings may have implications for the evaluation of PD.
Collapse
Affiliation(s)
| | - María-Del-Mar Guerra
- Department of Biochemistry, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Silvia Castellano
- Department of Metabolopathy, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
5
|
Sun M, Wang Y, Xu H, Shen Y, Liu B, Ma Y, Jiang C, Wang S, Li Q, Lu Y, Han F, Li T, Qin Y. Novel Hypochlorous Acid-Activated Near-Infrared Probe Monitors the Dynamic Changes of Myeloperoxidase Activity in Ischemic Brain. J Med Chem 2025; 68:5382-5399. [PMID: 40014579 DOI: 10.1021/acs.jmedchem.4c02431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Myeloperoxidase (MPO) contributes to the progression of ischemic damage. To fully understand MPO biology, highly sensitive and specific probes that can trace the activity of endogenous MPO fluxes are indispensable. Here, we developed two hypochlorous acid (HClO)-activated near-infrared probes to image MPO activity in a noninvasive manner. The probe MPO-NIR-II could track MPO-induced HClO in real time and in situ upon various stimuli with high sensitivity and specificity. Furthermore, MPO-NIR-II could monitor the MPO activity by in vivo fluorescence imaging and confocal laser scanning microscopy in mice with ischemic stroke. Moreover, a high-content screening system for MPO inhibitors was established by combining MPO-NIR-II with MPO-overexpressed cells and mouse brain slices with ischemic stroke, and the candidate compound AZD5904 was found to effectively attenuate ischemic brain injury. Overall, this work provides a versatile fluorescence tool that holds great promise for visualizing endogenous MPO fluxes of brain ischemia.
Collapse
Affiliation(s)
- Meiling Sun
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Yuting Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Huijun Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuting Shen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Bin Liu
- Department of Geriatrics, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Yuchen Ma
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chenchen Jiang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Supeng Wang
- The First Clinical Medical College of Nanjing Medical University, Nanjing 211166, China
| | - Qi Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yingmei Lu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Feng Han
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Tingyou Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yajuan Qin
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
6
|
Huang LY, Zhang YD, Chen J, Fan HD, Wang W, Wang B, Ma JY, Li PP, Pu HW, Guo XY, Shen JG, Qi SH. Maintaining moderate levels of hypochlorous acid promotes neural stem cell proliferation and differentiation in the recovery phase of stroke. Neural Regen Res 2025; 20:845-857. [PMID: 38886957 PMCID: PMC11433893 DOI: 10.4103/1673-5374.392889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/17/2023] [Accepted: 11/23/2023] [Indexed: 06/20/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202503000-00029/figure1/v/2024-06-17T092413Z/r/image-tiff It has been shown clinically that continuous removal of ischemia/reperfusion-induced reactive oxygen species is not conducive to the recovery of late stroke. Indeed, previous studies have shown that excessive increases in hypochlorous acid after stroke can cause severe damage to brain tissue. Our previous studies have found that a small amount of hypochlorous acid still exists in the later stage of stroke, but its specific role and mechanism are currently unclear. To simulate stroke in vivo, a middle cerebral artery occlusion rat model was established, with an oxygen-glucose deprivation/reoxygenation model established in vitro to mimic stroke. We found that in the early stage (within 24 hours) of ischemic stroke, neutrophils produced a large amount of hypochlorous acid, while in the recovery phase (10 days after stroke), microglia were activated and produced a small amount of hypochlorous acid. Further, in acute stroke in rats, hypochlorous acid production was prevented using a hypochlorous acid scavenger, taurine, or myeloperoxidase inhibitor, 4-aminobenzoic acid hydrazide. Our results showed that high levels of hypochlorous acid (200 μM) induced neuronal apoptosis after oxygen/glucose deprivation/reoxygenation. However, in the recovery phase of the middle cerebral artery occlusion model, a moderate level of hypochlorous acid promoted the proliferation and differentiation of neural stem cells into neurons and astrocytes. This suggests that hypochlorous acid plays different roles at different phases of cerebral ischemia/reperfusion injury. Lower levels of hypochlorous acid (5 and 100 μM) promoted nuclear translocation of β-catenin. By transfection of single-site mutation plasmids, we found that hypochlorous acid induced chlorination of the β-catenin tyrosine 30 residue, which promoted nuclear translocation. Altogether, our study indicates that maintaining low levels of hypochlorous acid plays a key role in the recovery of neurological function.
Collapse
Affiliation(s)
- Lin-Yan Huang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yi-De Zhang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jie Chen
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hai-Di Fan
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Department of Laboratory Medicine, Branch Hospital of Huai’an First People’s Hospital, Huai’an, Jiangsu Province, China
| | - Wan Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Bin Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ju-Yun Ma
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Peng-Peng Li
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hai-Wei Pu
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xin-Yian Guo
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jian-Gang Shen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Su-Hua Qi
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Wang H, Li K, Zhang L, Han T, Mu S, Song Y, Han Z, Zhao H, Liu X, Zhang H. Novel HOCl-Responsive Theranostic Prodrug for the Early Diagnosis and Therapy of Ischemic Stroke. J Med Chem 2025; 68:4951-4960. [PMID: 39937574 DOI: 10.1021/acs.jmedchem.4c03172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
In this work, a hypochlorous acid (HOCl)-responsive prodrug MB-R for diagnosis and therapy of ischemic stroke (IS) was constructed using the near-infrared fluorophore methylene blue linked to riluzole by the urea bond. MB-R exhibits good biocompatibility, fast response (<1 min), and high selectivity toward HOCl. MB-R was successfully utilized to visualize the HOCl levels, as well as the distribution of HOCl in the brains of IS mice to determine the progression of the disease. Meanwhile, the treatment with MB-R could reduce the cerebral infarction volume and improve the motor function in IS mice. Most importantly, MB-R could be utilized in the treatment of stroke through antioxidant, anti-inflammatory, and neuroprotective effects, further suggesting that riluzole was a potentially therapeutic agent for IS. Thus, this work paves the way for the development of intelligent theranostic agent for the early diagnosis and treatment of IS.
Collapse
Affiliation(s)
- Huayu Wang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Kemin Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Linjie Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Taihe Han
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Shuai Mu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Youwei Song
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Zehua Han
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Haiyu Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiaoyan Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| |
Collapse
|
8
|
Zhang S, Li R, Song M, Han J, Fan X. Exploration of M2 macrophage membrane as a biotherapeutic agent and strong synergistic therapeutic effects in ischemic stroke. J Control Release 2025; 378:476-489. [PMID: 39561947 DOI: 10.1016/j.jconrel.2024.11.033] [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: 07/18/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
The macrophage-derived membrane is widely applied in the targeting nanocarrier for its inflammatory tendency and long circulation ability due to the preservation of membrane protein. Few studies reported the application of macrophage membranes as biotherapeutic agents. To verify the ability of macrophage membrane as a biotherapeutic agent, ischemic stroke was selected as the model disease. Inspired by the features of macrophages infiltrating the ischemic core and the talent of M2 macrophages in modulating the inflammatory microenvironment, an M2 macrophage membrane (M2M)-disguised poly lactic-co-glycolic acid nanoparticles loaded with baicalin (BA) (M2M@BANPs) is developed. The results in vivo and in vitro indicate that M2M@BANPs could efficiently and actively target ischemic brain tissue and accumulate in microglia and neurons due to the coating of M2M. Furthermore, M2M and M2M@BANPs exhibit significant therapeutic effects in salvaging brain tissue damage and neurological functional recovery by reprogramming microglia from M1 to M2, reducing neutrophil infiltration and inhibiting neuronal apoptosis. Together, our fabrication provides a new insight and an applicative perspective for M2M in the therapy of ischemic stroke.
Collapse
Affiliation(s)
- Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jin Han
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
9
|
Huang Z, Liu Q, Guo Q, Gao J, Zhang L, Li L. Effects and mechanisms of Apelin in treating central nervous system diseases. Neuroscience 2025; 566:177-189. [PMID: 39681256 DOI: 10.1016/j.neuroscience.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Apelin, an endogenous ligand of G protein-coupled receptor APJ, is widely distributed in the central nervous system (CNS). It can be divided into such subtypes as Apelin-13, Apelin-17, and Apelin-36 as they have different amino acid structures. All Apelin is widely studied as an adipokine, showing a significant protective effect through regulating apoptosis, autophagy, oxidative stress, angiogenesis, inflammation, and other pathophysiological processes. As an adipokine, Apelin has been found to play a crucial role in cardiovascular disease development. In this paper, we reviewed the effects and mechanisms of Apelin in treating CNS diseases, such as neurotrauma, stroke, spinal cord injury, primary tumors, neurodegenerative diseases, psychiatric diseases, epilepsy, and pain.
Collapse
Affiliation(s)
- Zimeng Huang
- Medicine School, Qingdao University, 308 Ningxia Road, Shinan District, Qingdao 266071, China
| | - Qing Liu
- Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250021, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Qixuan Guo
- Department of Human Anatomy, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences and Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Luping Zhang
- Department of Human Anatomy, Binzhou Medical University, Yantai, Shandong, 264003, China.
| | - Liming Li
- Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266071, China.
| |
Collapse
|
10
|
Wang M, Xing S, Liu Y, An Z, Liu X, Liu T, Zhang H, Dai Y, Yang H, Wang Y, Wang Y. 2-Acetylacteoside improves recovery after ischemic stroke by promoting neurogenesis via the PI3K/Akt pathway. Free Radic Biol Med 2024; 225:415-429. [PMID: 39396583 DOI: 10.1016/j.freeradbiomed.2024.10.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024]
Abstract
Ischemic stroke induces adult neurogenesis in the subventricular zone (SVZ), even in elderly patients. Harnessing of this neuroregenerative response presents the therapeutic potential for post-stroke recovery. We found that phenylethanoid glycosides (PhGs) derived from Cistanche deserticola aid neural repair after stroke by promoting neurogenesis. Among these, 2-acetylacteoside had the most potent on the proliferation of neural stem cells (NSCs) in vitro. Furthermore, 2-acetylacteoside was shown to alleviate neural dysfunction by increase neurogenesis both in vivo and in vitro. RNA-sequencing analysis highlighted differentially expressed genes within the PI3K/Akt signaling pathway. The candidate target Akt was validated as being regulated by 2-acetylacteoside, which, in turn, enhanced the proliferation and differentiation of cultured NSCs after oxygen-glucose deprivation/reoxygenation (OGD/R), as evidenced by Western blot analysis. Subsequent analysis using cultured NSCs from adult subventricular zones (SVZ) confirmed that 2-acetylacteoside enhanced the expression of phosphorylated Akt (p-Akt), and its effect on NSC neurogenesis was shown to be dependent on the PI3K/Akt pathway. In summary, our findings elucidate for the first time the role of 2-acetylacteoside in enhancing neurological recovery, primarily by promoting neurogenesis via Akt activation following ischemic brain injury, which offers a novel strategy for long-term cerebrological recovery in ischemic stroke.
Collapse
Affiliation(s)
- Meng Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
| | - Songyu Xing
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
| | - Yang Liu
- ICU, Nanjing Gaochun People's Hospital, 53 Maoshan Road, Gaochun District, Nanjing, 211300, China
| | - Zongren An
- ICU, Nanjing Gaochun People's Hospital, 53 Maoshan Road, Gaochun District, Nanjing, 211300, China
| | - Xu Liu
- Qilu Medical University, Shandong, 255300, China
| | - Tao Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yifan Dai
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haiyuan Yang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China.
| | - Yu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Ying Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
11
|
Ning Y, Yuwen Zhou I, Caravan P. Quantitative in Vivo Molecular MRI. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407262. [PMID: 39279542 PMCID: PMC11530320 DOI: 10.1002/adma.202407262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Molecular magnetic resonance imaging (MRI) combines chemistry, chemical biology, and imaging techniques to track molecular events non-invasively. Quantitative molecular MRI aims to provide meaningful, reproducible numerical measurements of molecular processes or biochemical targets within the body. In this review, the classifications of molecular MRI probes based on their signal-generating mechanism and functionality are first described. From there, the primary considerations for in vitro characterization and in vivo validation of molecular MRI probes, including how to avoid pitfalls and biases are discussed. Then, recommendations on imaging acquisition protocols and analysis methods to establish quantitative relationships between MRI signal change induced by the probes and the molecular processes of interest are provided. Finally, several representative case studies are highlighted that incorporate these features. Quantitative molecular MRI is a multidisciplinary research area incorporating expertise in chemical biology, inorganic chemistry, molecular probes, imaging physics, drug development, pathobiology, and medicine. The purpose of this review is to provide guidance to chemists developing MR imaging probes and methods in terms of in vitro and in vivo validation to accelerate the translation of these new quantitative tools for non-invasive imaging of biological processes.
Collapse
Affiliation(s)
- Yingying Ning
- Spin-X Institute, School of Chemistry and Chemical Engineering, School of Biomedical Sciences and Engineering, State Key Laboratory of Luminescent Materials and Devices, Guangdong-Hong Kong-Macao Joint Laboratory of Optoelectronic and Magnetic Functional Materials, South China University of Technology, Guangzhou 510641, China
| | - Iris Yuwen Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
12
|
Zuo T, Shen C, Xie Z, Xu G, Wei F, Yang J, Zhu X, Hu Q, Zhao Z, Tang BZ, Cen Y. FRAME: flap endonuclease 1-engineered PAM module for precise and sensitive modulation of CRISPR/Cas12a trans-cleavage activity. Nucleic Acids Res 2024; 52:11884-11894. [PMID: 39315702 PMCID: PMC11514456 DOI: 10.1093/nar/gkae804] [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: 07/01/2024] [Revised: 08/27/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
CRISPR/Cas12a system, renowned for its precise recognition and efficient nucleic acid cleavage capabilities, has demonstrated remarkable performance in molecular diagnostics and biosensing. However, the reported Cas12a activity regulation methods often involved intricate CRISPR RNA (crRNA) structural adjustments or costly chemical modifications, which limited their applications. Here, we demonstrated a unique enzyme activity engineering strategy using flap endonuclease 1 (FEN1) to regulate the accessibility of the protospacer adjacent motif (PAM) module in the double-stranded DNA activator (FRAME). By identifying the three-base overlapping structure between the target inputs and substrate, FEN1 selectively cleaved and released the 5'-flap containing the 'TTTN' sequence, which triggered the secondary cleavage of FEN1 while forming a nicked PAM, ultimately achieving the sensitive switching of Cas12a's activity. The FRAME strategy exemplified the 'two birds with one stone' principle, as it not only precisely programmed Cas12a's activity but also simultaneously triggered isothermal cyclic amplification. Moreover, the FRAME strategy was applied to construct a sensing platform for detecting myeloperoxidase and miR-155, which demonstrated high sensitivity and specificity. Importantly, it proved its versatility in detecting multiple targets using a single crRNA without redesign. Collectively, the FRAME strategy opens up a novel avenue for modulating Cas12a's activity, promising immense potential in the realm of medical diagnostics.
Collapse
Affiliation(s)
- Tongshan Zuo
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chen Shen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhen Xie
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Guanhong Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Fangdi Wei
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jing Yang
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaolei Zhu
- Department of Neurology, Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Qin Hu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zheng Zhao
- Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Ben Zhong Tang
- Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Yao Cen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Clinical Medical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, Hunan Normal University, Ministry of Education, Changsha, Hunan 410081, China
| |
Collapse
|
13
|
Bu HF, Subramanian S, Chou PM, Liu F, Sun L, Geng H, Wang X, Liao J, Du C, Hu J, Tan SC, Nathan N, Yang GY, Tan XD. A novel mouse model of hepatocyte-specific apoptosis-induced myeloid cell-dominant sterile liver injury and repair response. Am J Physiol Gastrointest Liver Physiol 2024; 327:G499-G512. [PMID: 39104322 PMCID: PMC11482258 DOI: 10.1152/ajpgi.00005.2024] [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] [Received: 01/10/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024]
Abstract
Apoptosis, inflammation, and wound healing are critical pathophysiological events associated with various liver diseases. Currently, there is a lack of in vivo approaches to study hepatocyte apoptosis-induced liver injury and repair. To address this critical knowledge gap, we developed a unique genetically modified mouse model, namely, 3-Transgene (Tg) with inducible Hepatocyte-Specific Apoptosis Phenotype (3xTg-iHAP) in this study. The 3xTg-iHAP mice possess three transgenes including Alb-Cre, Rosa26-rtTA, and tetO-Fasl on a B6 background. These mice are phenotypically normal, viable, and fertile. After subcutaneous administration of a single dose of doxycycline (5 mg/kg, Dox) to 3xTg-iHAP mice, we observed a complete histological spectrum of sterile liver wound-healing responses: asymptomatic hepatocyte apoptosis at 8 h, necrotic liver injury and sterile inflammation at 48 h, followed by hepatocyte mitosis and regeneration within 7 days. During the injury phase, the mice exhibited an increase in the biomarkers of alanine aminotransferase (ALT), chemokine (C-X-C motif) ligand 1 (CXCL1), and IL-6 in peripheral blood, as well as α-smooth muscle actin (α-SMA) protein in liver tissues. Conversely, the mice displayed a decrease in these markers in the recovery phase. Remarkably, this model shows that the sterile liver injury following elevated hepatocyte apoptosis is associated with an increase in myeloid cells in the liver. Within 7 days post-Dox administration, the liver of Dox-treated 3xTg-iHAP mice displays a normal histological structure, indicating the completion of wound healing. Together, we established a novel mouse model of injury and regeneration induced by hepatocyte apoptosis. This tool provides a robust in vivo platform for studying the pathophysiology of sterile liver inflammation, regeneration, and new therapeutic interventions for liver diseases.NEW & NOTEWORTHY Bu et al. present a triple-transgenic mouse model, namely, 3xTg-iHAP mice that are engineered to explore hepatocyte apoptosis-triggered sterile liver injury and regeneration. This model demonstrates a full spectrum of liver wound-healing responses from asymptomatic apoptosis to injury, myeloid cell-dominant sterile inflammation, and repair after induction of hepatocyte-specific apoptosis. The robust nature of this model makes it an invaluable in vivo tool for studying sterile liver inflammation, regeneration, and new therapeutic strategies.
Collapse
Affiliation(s)
- Heng-Fu Bu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Saravanan Subramanian
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Pauline M Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Fangyi Liu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Leyu Sun
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Hua Geng
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Xiao Wang
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Jie Liao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Chao Du
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Joyce Hu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Stephanie C Tan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Nirmal Nathan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Xiao-Di Tan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| |
Collapse
|
14
|
Li P, Wang H, Niu C, Du X, Zhao M, Yang D, Jing W. The value of myeloperoxidase to high density lipoprotein ratio in predicting 90-day recurrence in patients with acute ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:107904. [PMID: 39116960 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107904] [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: 01/04/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
OBJECTIVE The ratio of myeloperoxidase to high-density lipoprotein (MPO/HDL) has become a novel inflammatory biomarker in the field of cardiovascular disease. MPO and HDL have been reported to be associated with inflammation and lipid metabolism after AIS. However, the effect of MPO/HDL on AIS recurrence has not been studied. We aimed to assess the value of MPO/HDL in predicting relapse 90 days after AIS. METHODS A total of 363 patients diagnosed with AIS were followed up for 90 days. Patients were assessed for recurrence within 90 days after AIS. Univariate and multivariate analyses were performed to determine the association between MPO/HDL and relapse within 90 days in AIS patients. The receiver operating characteristic curve (ROC) was used to compare the predictive value of MPO, HDL and MPO/HDL for recurrence at 90 days after AIS. RESULTS The proportion of recurrent stroke patients within 90 days was 6.61% (24/363). Recurrent stroke was associated with NIHSS, WBC, NEUT, UA, DD, Hcy, MPO, HDL, and MPO/HDL. After adjusting for potential confounders, the 90-day recurrence risk of AIS patients increased by 0.03 (P < 0.001) for each unit increase in MPO/HDL. The ROC curve constructed after correcting confounders found that compared with MPO(AUC=0.9698) and HDL(AUC=0.821), MPO/HDL showed the highest AUC value (AUC=0.9801), indicating that MPO/HDL levels had the highest predictive value for 90-day relapse in AIS patients. CONCLUSIONS MPO and MPO/HDL were independently associated with relapse within 90 days of AIS. MPO/HDL may be an independent predictor of 90-day relapse in AIS patients.
Collapse
Affiliation(s)
- Penghong Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Haobo Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Cailang Niu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xueqing Du
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Mina Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Debo Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wei Jing
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
15
|
Xu M, Feng G, Fang J. Microcapsules based on biological macromolecules for intestinal health: A review. Int J Biol Macromol 2024; 276:133956. [PMID: 39029830 DOI: 10.1016/j.ijbiomac.2024.133956] [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: 04/03/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Intestinal dysfunction is becoming increasingly associated with neurological and endocrine issues, raising concerns about its impact on world health. With the introduction of several breakthrough technologies for detecting and treating intestinal illnesses, significant progress has been made in the previous few years. On the other hand, traditional intrusive diagnostic techniques are expensive and time-consuming. Furthermore, the efficacy of conventional drugs (not capsules) is reduced since they are more likely to degrade before reaching their target. In this context, microcapsules based on different types of biological macromolecules have been used to encapsulate active drugs and sensors to track intestinal ailments and address these issues. Several biomacromolecules/biomaterials (natural protein, alginate, chitosan, cellulose and RNA etc.) are widely used for make microcapsules for intestinal diseases, and can significantly improve the therapeutic effect and reduce adverse reactions. This article systematically summarizes microencapsulated based on biomacromolecules material for intestinal health control and efficacy enhancement. It also discusses the application and mechanism research of microencapsulated biomacromolecules drugs in reducing intestinal inflammation, in addition to covering the preparation techniques of microencapsulated drug delivery systems used for intestinal health. Microcapsule delivery systems' limits and potential applications for intestinal disease diagnosis, treatment, and surveillance were highlighted.
Collapse
Affiliation(s)
- Minhui Xu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha 410128, Hunan, China
| | - Guangfu Feng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha 410128, Hunan, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha 410128, Hunan, China
| |
Collapse
|
16
|
Motlagh NJ, Wang C, Kim HH, Jun Y, Kim D, Lee S, Chen JW. Aging Intensifies Myeloperoxidase Activity after Ischemic Stroke. Aging Dis 2024; 15:2650-2664. [PMID: 39325942 PMCID: PMC11567251 DOI: 10.14336/ad.2023.1640] [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: 10/11/2023] [Accepted: 02/06/2024] [Indexed: 09/28/2024] Open
Abstract
Abnormally elevated oxidative stress underlies many diseases and contributes to aging. The myeloid enzyme myeloperoxidase (MPO) generates oxidative stress and contributes to damage after stroke. How aging changes MPO in stroke has not been studied. We aimed to determine the effects aging has on MPO and how these changes contribute to age-related differences in outcomes after ischemic stroke. To investigate tissue MPO activity we developed MPO Activatable Fluorescent Agent (MAFA). We found that aged mice exhibited worse neurological outcomes and higher mortality within the first few days after stroke. Accordingly, neuronal loss was higher in aged mice on day 3. MAFA imaging revealed that aged brains have markedly higher MPO activity that increased after stroke on day 3 compared to young adult brains. Correspondingly, we found more Iba1+ cells in aged brains compared to young adult brains before and after stroke. Interestingly, we found decreased percentage of MPO+ cells and lower MPO protein levels in aged on day 3, suggesting that most Iba1+ cells in aged mice have degranulated and secreted MPO in response to stroke. By day 10 MPO activity and Iba1+ cells decreased in both age groups, although MPO activity remained higher in aged mice. MPO inhibition in aged mice decreased MAFA signal and Iba1+ cells and improved neurobehavioral outcomes to near young adult stroke mice levels and improved mortality rate. While aging is an unmodifiable risk, by uncovering the connection between aging and MPO-related changes after stroke, new therapies can be developed to mitigate these adverse changes brought upon by aging.
Collapse
Affiliation(s)
- Negin Jalali Motlagh
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
| | - Cuihua Wang
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
| | - Hyung-Hwan Kim
- Stroke and Neurovascular Regulation Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | - Yonghyun Jun
- Stroke and Neurovascular Regulation Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | - Daeki Kim
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
| | - Seeun Lee
- Stroke and Neurovascular Regulation Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | - John W. Chen
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA.
| |
Collapse
|
17
|
Xu X, Chen M, Zhu D. Reperfusion and cytoprotective agents are a mutually beneficial pair in ischaemic stroke therapy: an overview of pathophysiology, pharmacological targets and candidate drugs focusing on excitotoxicity and free radical. Stroke Vasc Neurol 2024; 9:351-359. [PMID: 37832977 PMCID: PMC11420919 DOI: 10.1136/svn-2023-002671] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Stroke is the second-leading cause of death and the leading cause of disability in much of the world. In particular, China faces the greatest challenge from stroke, since the population is aged quickly. In decades of clinical trials, no neuroprotectant has had reproducible efficacy on primary clinical end points, because reperfusion is probably a necessity for neuroprotection to be clinically beneficial. Fortunately, the success of thrombolysis and endovascular thrombectomy has taken us into a reperfusion era of acute ischaemic stroke (AIS) therapy. Brain cytoprotective agents can prevent detrimental effects of ischaemia, and therefore 'freeze' ischaemic penumbra before reperfusion, extend the time window for reperfusion therapy. Because reperfusion often leads to reperfusion injury, including haemorrhagic transformation, brain oedema, infarct progression and neurological worsening, cytoprotective agents will enhance the efficacy and safety of reperfusion therapy by preventing or reducing reperfusion injuries. Therefore, reperfusion and cytoprotective agents are a mutually beneficial pair in AIS therapy. In this review, we outline critical pathophysiological events causing cell death within the penumbra after ischaemia or ischaemia/reperfusion in the acute phase of AIS, focusing on excitotoxicity and free radicals. We discuss key pharmacological targets for cytoprotective therapy and evaluate the recent advances of cytoprotective agents going through clinical trials, highlighting multitarget cytoprotective agents that intervene at multiple levels of the ischaemic and reperfusion cascade.
Collapse
Affiliation(s)
- Xiumei Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingyu Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongya Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Goh AR, Park J, Sim AY, Koo BN, Lee YH, Kim JY, Lee JE. Modulating monocyte-derived macrophage polarization in cerebral ischemic injury with hyperglycemia. Exp Neurol 2024; 378:114824. [PMID: 38777250 DOI: 10.1016/j.expneurol.2024.114824] [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: 02/04/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Ischemic stroke (IS), characterized by high mortality rate, occurs owing to diminished or blocked blood flow to the brain. Hyperglycemia (HG) is a major contributor to the risk of IS. HG induces augmented oxidative stress and Blood-Brain Barrier breakdown, which increases the influx of blood-derived myeloid cells into the brain parenchyma. In cerebral ischemia, infiltrating monocytes undergo differentiation into pro-inflammatory or anti-inflammatory macrophages, having a large effect on outcomes of ischemic stroke. In addition, interleukin-4 (IL-4) and interleukin-13 (IL-13) engage in post-ischemia repair by polarizing the infiltrating monocytes into an anti-inflammatory phenotype. In this study, we aimed to determine the effect of phenotypic polarization of monocyte-derived macrophages on the prognosis of IS with HG (HG-IS). We first established a hyperglycemic mouse model using streptozotocin (150 mg/kg) and induced transient middle cerebral artery occlusion. We observed that blood-brain barrier permeability increased in HG-IS mice, as per two-photon live imaging and Evans blue staining. We also confirmed the increased infiltration of monocyte-derived macrophages and the downregulation of anti-inflammatory macrophages related to tissue remodeling after inflammation in HG-IS mice through immunohistochemistry, western blotting, and flow cytometry. We observed phenotypic changes in monocyte-derived macrophages, alleviated infarct volume, and improved motor function in HG-IS mice treated with IL-4 and IL-13. These findings suggest that the modulation of phenotypic changes in monocyte-derived macrophages following IS in hyperglycemic mice may influence ischemic recovery.
Collapse
Affiliation(s)
- A Ra Goh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joohyun Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - A Young Sim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea; Interdisciplinary Program of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Li P, Niu C, Du X, Zhao M, Wang H, Yang D, Li Y, Jing W. Myeloperoxidase to high-density lipoprotein ratio: Potential predictor of severity and outcome in patients with acute ischemic stroke. Brain Res 2024; 1833:148883. [PMID: 38521161 DOI: 10.1016/j.brainres.2024.148883] [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: 02/02/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVE As a new marker of inflammation and lipid metabolism, the ratio of myeloperoxidase to high density lipoprotein (MPO/HDL) has been reported in the field of cardiovascular disease. However, the effect of MPO/HDL on acute ischemic stroke (AIS) is not clear. The purpose of this study was to explore the prognostic value of MPO/HDL level in patients with AIS. METHODS This study conducted a retrospective analysis of 363 patients diagnosed with AIS. Stroke severity was assessed by National Institutes of Health Stroke Scale (NIHSS). The short-term functional outcome was evaluated with modified Rankin Scale (mRS) 90 days after admission. Spearman correlation analysis was used to evaluate the correlation between MPO/HDL and NIHSS scores. The predictive value of MPO, HDL and MPO/HDL to AIS was evaluated by receiver operating characteristic curve (ROC). RESULTS The level of MPO/HDL in patients with NIHSS score ≥ 4 was significantly higher than that in patients with NIHSS score < 4 (P < 0.001). MPO and MPO/HDL were positively correlated with NIHSS score (P < 0.001), while HDL was negatively correlated with NIHSS score (P < 0.001). During 90-day follow-up, multivariate Logistic regression analysis showed that increased MPO/HDL levels were associated with 90-day functional outcomes. ROC showed that compared with MPO and HDL, MPO/HDL had the highest predictive value for 90-day functional prognosis in patients with AIS (AUC = 0.9825). CONCLUSION The level of serum MPO/HDL may be potential prognostic biomarker in AIS 90 days.
Collapse
Affiliation(s)
- Penghong Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China
| | - Cailang Niu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China
| | - Xueqing Du
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China
| | - Mina Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China
| | - Haobo Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China
| | - Debo Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China
| | - Yuan Li
- Shanxi Cardiovascular Hospital, Taiyuan 030032, China
| | - Wei Jing
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032,China.
| |
Collapse
|
20
|
Parsi S, Zhu C, Motlagh NJ, Kim D, Küllenberg EG, Kim HH, Gillani RL, Chen JW. Basic Science of Neuroinflammation and Involvement of the Inflammatory Response in Disorders of the Nervous System. Magn Reson Imaging Clin N Am 2024; 32:375-384. [PMID: 38555147 PMCID: PMC10987041 DOI: 10.1016/j.mric.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Neuroinflammation is a key immune response observed in many neurologic diseases. Although an appropriate immune response can be beneficial, aberrant activation of this response recruits excessive proinflammatory cells to cause damage. Because the central nervous system is separated from the periphery by the blood-brain barrier (BBB) that creates an immune-privileged site, it has its own unique immune cells and immune response. Moreover, neuroinflammation can compromise the BBB causing an influx of peripheral immune cells and factors. Recent advances have brought a deeper understanding of neuroinflammation that can be leveraged to develop more potent therapies and improve patient selection.
Collapse
Affiliation(s)
- Sepideh Parsi
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cindy Zhu
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Negin Jalali Motlagh
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daeki Kim
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Enrico G Küllenberg
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyung-Hwan Kim
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca L Gillani
- Department of Neurology, Neuroimmunology and Neuro-Infectious Diseases Division, Massachusetts Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John W Chen
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Duan M, Xu Y, Li Y, Feng H, Chen Y. Targeting brain-peripheral immune responses for secondary brain injury after ischemic and hemorrhagic stroke. J Neuroinflammation 2024; 21:102. [PMID: 38637850 PMCID: PMC11025216 DOI: 10.1186/s12974-024-03101-y] [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: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
The notion that the central nervous system is an immunologically immune-exempt organ has changed over the past two decades, with increasing evidence of strong links and interactions between the central nervous system and the peripheral immune system, both in the healthy state and after ischemic and hemorrhagic stroke. Although primary injury after stroke is certainly important, the limited therapeutic efficacy, poor neurological prognosis and high mortality have led researchers to realize that secondary injury and damage may also play important roles in influencing long-term neurological prognosis and mortality and that the neuroinflammatory process in secondary injury is one of the most important influences on disease progression. Here, we summarize the interactions of the central nervous system with the peripheral immune system after ischemic and hemorrhagic stroke, in particular, how the central nervous system activates and recruits peripheral immune components, and we review recent advances in corresponding therapeutic approaches and clinical studies, emphasizing the importance of the role of the peripheral immune system in ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Mingxu Duan
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya Xu
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanshu Li
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yujie Chen
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
22
|
Chen S, Pan J, Gong Z, Wu M, Zhang X, Chen H, Yang D, Qi S, Peng Y, Shen J. Hypochlorous acid derived from microglial myeloperoxidase could mediate high-mobility group box 1 release from neurons to amplify brain damage in cerebral ischemia-reperfusion injury. J Neuroinflammation 2024; 21:70. [PMID: 38515139 PMCID: PMC10958922 DOI: 10.1186/s12974-023-02991-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 12/11/2023] [Indexed: 03/23/2024] Open
Abstract
Myeloperoxidase (MPO) plays critical role in the pathology of cerebral ischemia-reperfusion (I/R) injury via producing hypochlorous acid (HOCl) and inducing oxidative modification of proteins. High-mobility group box 1 (HMGB1) oxidation, particularly disulfide HMGB1 formation, facilitates the secretion and release of HMGB1 and activates neuroinflammation, aggravating cerebral I/R injury. However, the cellular sources of MPO/HOCl in ischemic brain injury are unclear yet. Whether HOCl could promote HMGB1 secretion and release remains unknown. In the present study, we investigated the roles of microglia-derived MPO/HOCl in mediating HMGB1 translocation and secretion, and aggravating the brain damage and blood-brain barrier (BBB) disruption in cerebral I/R injury. In vitro, under the co-culture conditions with microglia BV cells but not the single culture conditions, oxygen-glucose deprivation/reoxygenation (OGD/R) significantly increased MPO/HOCl expression in PC12 cells. After the cells were exposed to OGD/R, MPO-containing exosomes derived from BV2 cells were released and transferred to PC12 cells, increasing MPO/HOCl in the PC12 cells. The HOCl promoted disulfide HMGB1 translocation and secretion and aggravated OGD/R-induced apoptosis. In vivo, SD rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) plus different periods of reperfusion. Increased MPO/HOCl production was observed at the reperfusion stage, accomplished with enlarged infarct volume, aggravated BBB disruption and neurological dysfunctions. Treatment of MPO inhibitor 4-aminobenzoic acid hydrazide (4-ABAH) and HOCl scavenger taurine reversed those changes. HOCl was colocalized with cytoplasm transferred HMGB1, which was blocked by taurine in rat I/R-injured brain. We finally performed a clinical investigation and found that plasma HOCl concentration was positively correlated with infarct volume and neurological deficit scores in ischemic stroke patients. Taken together, we conclude that ischemia/hypoxia could activate microglia to release MPO-containing exosomes that transfer MPO to adjacent cells for HOCl production; Subsequently, the production of HOCl could mediate the translocation and secretion of disulfide HMGB1 that aggravates cerebral I/R injury. Furthermore, plasma HOCl level could be a novel biomarker for indexing brain damage in ischemic stroke patients.
Collapse
Affiliation(s)
- Shuang Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jingrui Pan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhe Gong
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiaoni Zhang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hansen Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Dan Yang
- Department of Chemistry, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Suhua Qi
- Medical and Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China.
| | - Ying Peng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Medical and Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
23
|
Tang W, Li F, Huang R, Liu P. Causal relationship between levels of myeloperoxidase and obstructive sleep apnea: a bidirectional two-sample Mendelian randomization study. Front Neurol 2023; 14:1305580. [PMID: 38156091 PMCID: PMC10753018 DOI: 10.3389/fneur.2023.1305580] [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: 10/02/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Background Several observational studies have investigated the association between myeloperoxidase (MPO) and obstructive sleep apnea (OSA). However, the nature of this relationship remains uncertain due to potential selection and confounding biases. To resolve this, we conducted a bidirectional two-sample Mendelian randomization (MR) study to scrutinize the causal relationship between MPO and OSA. Methods Instrumental variables (IVs) for OSA were sourced from the publicly available FinnGen dataset, encompassing 38,998 OSA cases and 336,659 controls. Data on MPO were sourced from a study of 21,758 individuals conducted by the European Bioinformatics Institute (EBI). The primary MR analysis utilized the inverse-variance weighted (IVW) method, with MR-Egger intercept and leave-one-out methods assessing pleiotropy and Cochran's Q test determining heterogeneity. Results The IVW analysis indicated a causal relationship between heightened MPO levels and an increased incidence of OSA. Individuals with elevated MPO levels manifested a higher propensity to develop OSA, exhibiting an odds ratio (OR) of 1.075 and a 95% confidence interval (CI) of 1.011-1.143 (p = 0.021). Conversely, the reciprocal analysis unveiled no significant association between OSA and heightened MPO levels (p = 0.643). No directional pleiotropy was identified through the MR-Egger intercept test (p > 0.05). Conclusion Our study provides evidence of an association between elevated MPO levels and an increased incidence of OSA. However, OSA does not necessarily lead to elevated MPO levels. When patients present with high MPO levels, screening for OSA may be advisable, considering their clinical characteristics.
Collapse
Affiliation(s)
- Weihua Tang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
- Department of Radiology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Fang Li
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Rui Huang
- Department of Cardiology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Peijun Liu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| |
Collapse
|
24
|
Zhu C, Li Y, Deng Q, Liu X, Xia Q, Zhong L, Xia Z, ShanZhou Q, Lei J, Zhu J. Myeloperoxidase-Sensitive Magnetic Resonance Imaging Assesses Inflammatory Activation State in Experimental Mouse Acute Gout. J Magn Reson Imaging 2023; 58:1714-1722. [PMID: 37078554 DOI: 10.1002/jmri.28752] [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: 01/24/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND A novel myeloperoxidase-activatable manganese-based (MPO-Mn) MRI probe may enable the activation state of inflammatory foci to be detected and monitored noninvasively. PURPOSE To evaluate the inflammatory response in a mouse model of acute gout using MPO as an imaging biomarker and a potential therapeutic target. STUDY TYPE Prospective. ANIMAL MODEL A total of 40 male Swiss mice with monosodium urate crystals induced acute gout. FIELD STRENGTH/SEQUENCE A 3.0 T/T1-weighted imaging with 2D fast spoiled gradient recalled echo and T2-weighted imaging with fast recovery fast spin-echo sequences. ASSESSMENT The difference in contrast-to-noise ratio between left hind limb (lesion) and right hind limb (internal reference) (ΔCNR), and normalized signal-to-noise ratio (nSNR) on the right hind limb were calculated and compared. The expression level and activity of myeloperoxidase (MPO) were analyzed using western blotting and spectrophotometric quantitation activity assay. MPO-positive cell infiltration and lesion volume were evaluated using immunofluorescence staining and T2-weighted images, respectively. STATISTICAL TESTS Student's t test. A P-value less than 0.05 was considered to be statistically significant. RESULTS MPO-Mn resulted in a significantly higher ΔCNR than Gd-DTPA (22.54 ± 1.86 vs. 13.90 ± 2.22) but lower nSNR on the reference right hind limb (1.08 ± 0.07 vs. 1.21 ± 0.08). Compared to the nontreatment group, MPO-inhibition resulted in a significantly reduced contrast enhancement at the lesion (17.81 ± 1.58 vs. 22.96 ± 3.12), which was consistent with a remission of the inflammatory response, as evidenced by a substantial reduction of lesion volume (0.55 ± 0.16 mm3 /g vs. 1.14 ± 0.15 mm3 /g), myeloperoxidase expression level (0.98 ± 0.09 vs. 1.48 ± 0.19) and activity (0.75 ± 0.12 vs. 1.12 ± 0.07), and inflammatory cell recruitment. DATA CONCLUSION MPO-Mn MRI has potential to evaluate the activation state of inflammatory foci in the experimental model of acute gout. EVIDENCE LEVEL 1. TECHNICAL EFFICACY Stage 1.
Collapse
Affiliation(s)
- Chunrong Zhu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
- Department of Oncology Ward 2, Chengdu Third People's Hospital, Chengdu, Sichuan, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yunhe Li
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiao Deng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
| | - Xinxin Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
| | - Qian Xia
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
| | - Lei Zhong
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
| | - Zhiyang Xia
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
| | - Qiyue ShanZhou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
- Department of Hematology and Oncology, Chongzhou People's Hospital, Chongzhou, Sichuan, China
| | - Jun Lei
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiang Zhu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan, China
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
25
|
Liu Y, Xiang C, Que Z, Li C, Wang W, Yin L, Chu C, Zhou Y. Neutrophil heterogeneity and aging: implications for COVID-19 and wound healing. Front Immunol 2023; 14:1201651. [PMID: 38090596 PMCID: PMC10715311 DOI: 10.3389/fimmu.2023.1201651] [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: 04/06/2023] [Accepted: 08/02/2023] [Indexed: 12/18/2023] Open
Abstract
Neutrophils play a critical role in the immune response to infection and tissue injury. However, recent studies have shown that neutrophils are a heterogeneous population with distinct subtypes that differ in their functional properties. Moreover, aging can alter neutrophil function and exacerbate immune dysregulation. In this review, we discuss the concept of neutrophil heterogeneity and how it may be affected by aging. We then examine the implications of neutrophil heterogeneity and aging for COVID-19 pathogenesis and wound healing. Specifically, we summarize the evidence for neutrophil involvement in COVID-19 and the potential mechanisms underlying neutrophil recruitment and activation in this disease. We also review the literature on the role of neutrophils in the wound healing process and how aging and neutrophil heterogeneity may impact wound healing outcomes. Finally, we discuss the potential for neutrophil-targeted therapies to improve clinical outcomes in COVID-19 and wound healing.
Collapse
Affiliation(s)
| | | | | | | | - Wen Wang
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| | - Lijuan Yin
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| | - Chenyu Chu
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| | - Yin Zhou
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Medical Cosmetic Center, Chengdu Second People's Hospital; Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Ma W, Sun R, Tang L, Li Z, Lin L, Mai Z, Chen G, Yu Z. Bioactivable STING Nanoagonists to Synergize NIR-II Mild Photothermal Therapy Primed Robust and Long-Term Anticancer Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303149. [PMID: 37691545 DOI: 10.1002/adma.202303149] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Pharmacological activation of the stimulator of interferon genes (STING) pathway has become a promising strategy for cancer immunotherapy. However, the insufficient tumorous accumulation, rapid clearance, and short duration of drug efficacy in the tumor microenvironment of small structural STING agonists greatly compromise the therapeutic efficacy. Herein, a tumorous extracellular matrix (ECM) is presented anchoring STING agonist-based photoimmunothernostic nanomedicine (SAPTN) that can be activated by mild-temperature photothermal therapy (mild PTT) induced neutrophilic inflammation. The SAPTN owns second window near-infrared (NIR-II) photonics properties fitting for NIR-II fluorescence and photoacoustic imaging-guided cancer therapy. The aggregates SAPTN targeting to the ECM provide slow and continuous release of potent STING agonists diABZIs. The mild PTT and long-lasting STING agonists released in the ECM synergistically prime systematic, robust, and long-term anticancer immunity. In a tumor model, this approach leads to complete tumor eradication in about 100% of mice with orthotopic breast tumors, and the mice regained tumor-free survival of at least 2 months. In addition, the immune-mediated abscopal effect shows inhibition of the distant solid tumor growth by intratumoral administration of SAPTN with laser irradiation. Overall, this approach represents a generalized photoactivable nanomedicine to prime anticancer immunity for improved cancer theranostics.
Collapse
Affiliation(s)
- Wen Ma
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Rui Sun
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Longguang Tang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Zibo Li
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Ling Lin
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Ziyi Mai
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Gui Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
27
|
Kim J, Kang S, Choi MH, Park S, Nam SH, Park JU, Lee Y. Zwitterionic polymer on silicone implants inhibits the bacteria-driven pathogenic mechanism and progress of breast implant-associated anaplastic large cell lymphoma. Acta Biomater 2023; 171:378-391. [PMID: 37683967 DOI: 10.1016/j.actbio.2023.09.003] [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: 04/21/2023] [Revised: 08/22/2023] [Accepted: 09/03/2023] [Indexed: 09/10/2023]
Abstract
Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) occurs in the capsule surrounding breast implants. Malignant transformation of T cells by bacteria-driven chronic inflammation may be underlying BIA-ALCL mechanism. Here, we covalently grafted 2-methacryloyloxyethyl phosphorylcholine (MPC)-based polymers on a silicone surface and examined its effects against BIA-ALCL pathogenesis. MPC grafting strongly inhibited the adhesion of bacteria and bacteria-causing inflammation. Additionally, cancer T cell proliferation and capsule-derived fibroblast-cancer cell communication were effectively inhibited by MPC grafting. We further demonstrated the effect of MPC against the immune responses causing BIA-ALCL around human silicone implants in micro-pigs. Finally, we generated a xenograft anaplastic T cell lymphoma mouse model around the silicone implants and demonstrated that MPC grafting could effectively inhibit the lymphoma progression. This study is the first to show that bacteria-driven induction and progression of BIA-ALCL can be effectively inhibited by surface modification of implants. STATEMENT OF SIGNIFICANCE: Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is a major concern in the field of plastic and reconstructive surgery. In this study, we demonstrate strong inhibitory effect of zwitterionic polymer grafting on BIA-ALCL pathogenesis and progression, induced by bacterial infection and inflammation, both in vitro and in vivo. This study provides a molecular basis for the development of novel breast implants that can prevent various potential complications such as excessive capsular contracture, breast implant illness, and BIA-ALCL incidence, as well as for expanding the biomedical applications of zwitterionic polymers.
Collapse
Affiliation(s)
- Jungah Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Sunah Kang
- Department of Chemistry, College of Natural Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Min-Ha Choi
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 5 Gil 20, Boramae-ro, Dongjak-gu, Seoul 07061, Republic of Korea
| | - Sohyun Park
- Department of Chemistry, College of Natural Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - So Hee Nam
- College of Pharmacy, Dongduk Women's University, 60 Hwarang-ro 13-gil, Seongbuk-gu, Seoul 02748, Republic of Korea
| | - Ji-Ung Park
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 5 Gil 20, Boramae-ro, Dongjak-gu, Seoul 07061, Republic of Korea; Institute of Medical and Biological Engineering, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Yan Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
28
|
Kim D, Lee DW, Yoon G, Jeong EK, Choi MS, Lee HC, Park YS, Chung CP, Lee JY, Park YJ. Therapeutic Effect of HDAC5 Binding and Cell Penetrating Peptide for the Treatment of Inflammatory Bowel Disease. Tissue Eng Regen Med 2023; 20:965-979. [PMID: 37589886 PMCID: PMC10519921 DOI: 10.1007/s13770-023-00572-7] [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: 06/03/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is an incurable disease that negatively influences the quality of life of patients. Current and emerging therapies target proinflammatory cytokines and/or receptors to downregulate proinflammatory responses, but insufficient remission requires other therapeutic agents. Herein, we report that the synthetic anti-inflammatory peptide 15 (SAP15) is capable of cell penetration and anti-inflammatory activity in human macrophages. METHODS SAP15 was labeled with fluorescence and administered to human leukemia monocytic cells (THP-1) cells for cell penetration analysis. Using biolayer interferometry analysis, the binding affinity of SAP15 with histone deacetylase 5 (HDAC5) was measured. SAP15-treated THP-1 cells were analyzed by protein phosphorylation assay, flow cytometry, and enzyme-linked immunosorbent assay (ELISA). In addition, in vivo analysis of the therapeutic effect on IBD was observed in a dextran sulfate sodium (DSS)-induced model. Samples from SAP15-treated mice were analyzed at both the macroscopic and microscopic levels using ELISA, myeloperoxidase (MPO) assays, and histological evaluations. RESULTS SAP15 was internalized within the cytosol and nucleus of THP-1 cells and bound to the HDAC5 protein. SAP15-treated macrophages were assessed for protein phosphorylation and showed inhibited phosphorylation of HDAC5 and other immune-related proteins, which led to increased M2-like macrophage markers and decreased M1-like macrophage markers and tumor necrosis factor-α and interleukin-6 cytokine levels. The SAP15 treatment on IBD model showed significant recovery of colon length. Further histological analysis of colon demonstrated the therapeutic effect of SAP15 on mucosal layer. Moreover, proinflammatory cytokine levels and MPO activity from the plasma show that SAP15 is effective in reduced proinflammatory responses. CONCLUSION These findings suggest that SAP15 is a novel peptide with a novel cell-penetrating peptide with anti-inflammatory property that can be used as a therapeutic agent for IBD and other inflammatory diseases.
Collapse
Affiliation(s)
- Deogil Kim
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
| | - Dong Woo Lee
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
- Department of Dental Regenerative Biotechnology and Dental Research Institute, School of Dentistry, Seoul National University, #403 Biomaterial Research Building, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Gookjin Yoon
- Department of Dental Regenerative Biotechnology and Dental Research Institute, School of Dentistry, Seoul National University, #403 Biomaterial Research Building, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Eui Kyun Jeong
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
| | - Moon Sil Choi
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
| | - Hoo Cheol Lee
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
| | - Yoon Shin Park
- Department of Biological Sciences and Biotechnology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Chong Pyung Chung
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
| | - Jue-Yeon Lee
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea
| | - Yoon Jeong Park
- Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), Seoul, 03127, Republic of Korea.
- Department of Dental Regenerative Biotechnology and Dental Research Institute, School of Dentistry, Seoul National University, #403 Biomaterial Research Building, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
29
|
de Sousa MN, da Anunciação LF, de Freitas PLZ, Ricardo-da-Silva FY, Moreira LFP, Correia CJ, Breithaupt-Faloppa AC. Evaluation of the therapeutic effects of oestradiol on the systemic inflammatory response and on lung injury caused by the occlusion of the proximal descending aorta in male rats. Eur J Cardiothorac Surg 2023; 64:ezad253. [PMID: 37410160 DOI: 10.1093/ejcts/ezad253] [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] [Received: 02/24/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/07/2023] Open
Abstract
OBJECTIVES Ischaemia and reperfusion-induced microvascular dysfunction is a serious problem encountered during a variety surgical procedures, leading to systemic inflammation and affecting remote organs, specially the lungs. 17β-Oestradiol reduces pulmonary repercussions from various acute lung injury forms. Here, we focused on the 17β-oestradiol therapeutic effects after aortic ischaemia and reperfusion (I/R) by evaluating lung inflammation. METHODS Twenty-four Wistar rats were submitted to I/R by insufflation of a 2-F catheter in thoracic aorta for 20 min. Reperfusion took 4 h and 17β-oestradiol (280 µg/kg, i.v.) was administered after 1 h of reperfusion. Sham-operated rats were controls. Bronchoalveolar lavage was performed and lung samples were prepared for histopathological analysis and tissue culture (explant). Interleukin (IL)-1β, IL-10 and tumour necrosis factor-α were quantified. RESULTS After I/R, higher number of leukocytes in bronchoalveolar lavage were reduced by 17β-oestradiol. The treatment also decreased leukocytes in lung tissue. I/R increased lung myeloperoxidase expression, with reduction by 17β-oestradiol. Serum cytokine-induced neutrophil chemoattractant 1 and IL-1β increased after I/R and 17β-oestradiol decreased cytokine-induced neutrophil chemoattractant 1. I/R increased IL-1β and IL-10 in lung explants, reduced by 17β-oestradiol. CONCLUSIONS Our results showed that 17β-oestradiol treatment performed in the period of reperfusion, modulated the systemic response and the lung repercussions of I/R by thoracic aortic occlusion. Thus, we can suggest that 17β-oestradiol might be a supplementary approach leading the lung deterioration after aortic clamping in surgical procedures.
Collapse
Affiliation(s)
- Marcelo Nunes de Sousa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Lucas Ferreira da Anunciação
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Pedro Luiz Zonta de Freitas
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Yamamoto Ricardo-da-Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Cristiano Jesus Correia
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
Hua M, Chen WY, Wang LH, Zou XH, Mao LL. The value of serum Lp-PLA2 combined with MPO in the diagnosis of cerebral infarction caused by large artery atherosclerosis. Clin Neurol Neurosurg 2023; 232:107899. [PMID: 37467579 DOI: 10.1016/j.clineuro.2023.107899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/25/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
OBJECTIVE To explore the value of serum lipoprotein-associated phospholipase A2(Lp-PLA2)combined with myeloperoxidase(MPO)for the diagnosis of large artery atherosclerosis(LAA) cerebral infarction. METHODS Baseline data were collected from patients with first-ever acute cerebral infarction, serum Lp-PLA2 and MPO levels were measured. The etiology of cerebral infarction was classified according to the Chinese Ischemic Stroke Subtype Classification Standard. The risk factors associated with LAA cerebral infarction were identified by univariate and multivariate regression analysis. The diagnostic value of serum Lp-PLA2 and MPO for LAA cerebral infarction was assessed by the area under the receiver-operating characteristic (ROC) curve. RESULTS Overall 368 patients were involved, 148 patients (40.22 %) were LAA. The serum La-PLA2 and MPO levels were higher in the LAA group than those in non-LAA group (23.06 ± 3.39 ng/mL versus 17.48 ± 3.26 ng/mL; 93.60 ± 9.58 ng/mL versus 75.98 ± 15.53 ng/mL; P < 0.001 for both). Multivariate analysis showed that elevated levels of serum Lp-PLA2 (OR 1.742, 95 %CI 1.499-2.025; P < 0.001) and MPO (OR 1.060, 95 % CI 1.026-1.096; P = 0.001) were the independent risk factors of LAA cerebral infarction. The area under curve of the serum Lp-PLA2 combined with MPO for the diagnosis of LAA cerebral infarction was 0.896 [0.866 ∼ 0.927] (P < 0.001). CONCLUSION Serum Lp-PLA2 combined with MPO could be valued as a predictor of acute cerebral infarction caused by large artery atherosclerosis.
Collapse
Affiliation(s)
- Min Hua
- Department of Neurology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213002, Jiangsu Province, China
| | - Wen-Ya Chen
- Department of Neurology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213002, Jiangsu Province, China
| | - Li-Hui Wang
- Department of Neurology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213002, Jiangsu Province, China
| | - Xiao-Hua Zou
- Department of Neurology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213002, Jiangsu Province, China
| | - Lun-Lin Mao
- Department of Neurology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213002, Jiangsu Province, China.
| |
Collapse
|
31
|
King RM, Gounis MJ, Schmidt EJ, Leporati A, Gale EM, Bogdanov AA. Molecular Magnetic Resonance Imaging of Aneurysmal Inflammation Using a Redox Active Iron Complex. Invest Radiol 2023; 58:656-662. [PMID: 36822678 PMCID: PMC10401906 DOI: 10.1097/rli.0000000000000960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
OBJECTIVES Inflammation plays a key role in driving brain aneurysmal instability and rupture, but clinical tools to noninvasively differentiate between inflamed and stable aneurysms are lacking. We hypothesize that imaging oxidative changes in the aneurysmal microenvironment driven by myeloid inflammatory cells may represent a noninvasive biomarker to evaluate rupture risk. In this study, we performed initial evaluation of the oxidatively activated probe Fe-PyC3A as a tool for magnetic resonance imaging (MRI) of inflammation in a rabbit model of saccular aneurysm. MATERIALS AND METHODS The difference in longitudinal relaxivity ( r1 ) in reduced and oxidized states of Fe-PyC3A was measured in water and blood plasma phantoms at 3 T. A rabbit saccular aneurysm model was created by endovascular intervention/elastinolysis with subsequent decellularization in situ. Rabbits were imaged at 4 weeks (n = 4) or 12 weeks (n = 4) after aneurysmal induction, when luminal levels of inflammation reflected by the presence of myeloperoxidase positive cells are relatively high and low, respectively, using a 3 T clinical scanner. Both groups were imaged dynamically using a 2-dimensional T1-weighted fast field echo pulse MRI sequence before and up to 4 minutes postinjection of Fe-PyC3A. Dynamic imaging was then repeated after an injection of gadobutrol (0.1 mmol/kg) as negative control probe. Rabbits from the 12-week aneurysm group were also imaged before and 20 minutes and 3 hours after injection of Fe-PyC3A using an axial respiratory gated turbo-spin echo (TSE) pulse sequence with motion-sensitized driven equilibrium (MSDE) preparation. The MSDE/TSE imaging was repeated before, immediately after dynamic acquisition (20 minutes postinjection), and 3 hours after injection of gadobutrol. Aneurysmal enhancement ratios (ERs) were calculated by dividing the postinjection aneurysm versus skeletal muscle contrast ratio by the preinjection contrast ratio. After imaging, the aneurysms were excised and inflammatory infiltrate was characterized by fluorometric detection of myeloperoxidase activity and calprotectin immunostaining, respectively. RESULTS In vitro relaxometry showed that oxidation of Fe-PyC3A by hydrogen peroxide resulted in a 15-fold increase of r1 at 3 T. Relaxometry in the presence of blood plasma showed no more than a 10% increase of r1 , indicating the absence of strong interaction of Fe-PyC3A with plasma proteins. Dynamic imaging with Fe-PyC3A generated little signal enhancement within the blood pool or adjacent muscle but did generate a transient increase in aneurysmal ER that was significantly greater 4 weeks versus 12 weeks after aneurysm induction (1.6 ± 0.30 vs 1.2 ± 0.03, P < 0.05). Dynamic imaging with gadobutrol generated strong aneurysmal enhancement, but also strong enhancement of the blood and muscle resulting in smaller relative ER change. In the 12-week group of rabbits, MSDE/TSE imaging showed that ER values measured immediately after dynamic MRI (20 minutes postinjection) were significantly higher ( P < 0.05) in the case of Fe-PyC3A (1.25 ± 0.06) than for gadobutrol injection (1.03 ± 0.03). Immunohistochemical corroboration using anticalprotectin antibody showed that leukocyte infiltration into the vessel walls and luminal thrombi was significantly higher in the 4-week group versus 12-week aneurysms (123 ± 37 vs 18 ± 7 cells/mm 2 , P < 0.05). CONCLUSIONS Magnetic resonance imaging using Fe-PyC3A injection in dynamic or delayed acquisition modes was shown to generate a higher magnetic resonance signal enhancement in aneurysms that exhibit higher degree of inflammation. The results of our pilot experiments support further evaluation of MRI using Fe-PyC3A as a noninvasive marker of aneurysmal inflammation.
Collapse
Affiliation(s)
- Robert M King
- From the Department of Radiology and New England Center for Stroke Research, UMASS Chan Medical School, Worcester
| | - Matthew J Gounis
- From the Department of Radiology and New England Center for Stroke Research, UMASS Chan Medical School, Worcester
| | - Eric J Schmidt
- From the Department of Radiology and New England Center for Stroke Research, UMASS Chan Medical School, Worcester
| | - Anita Leporati
- From the Department of Radiology and New England Center for Stroke Research, UMASS Chan Medical School, Worcester
| | - Eric M Gale
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown
| | - Alexei A Bogdanov
- From the Department of Radiology and New England Center for Stroke Research, UMASS Chan Medical School, Worcester
| |
Collapse
|
32
|
Meade CS, Bell RP, Towe SL, Lascola CD, Al‐Khalil K, Gibson MJ. Cocaine use is associated with cerebral white matter hyperintensities in HIV disease. Ann Clin Transl Neurol 2023; 10:1633-1646. [PMID: 37475160 PMCID: PMC10502656 DOI: 10.1002/acn3.51854] [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: 04/10/2023] [Revised: 06/16/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND White matter hyperintensities (WMH), a marker of cerebral small vessel disease and predictor of cognitive decline, are observed at higher rates in persons with HIV (PWH). The use of cocaine, a potent central nervous system stimulant, is disproportionately common in PWH and may contribute to WMH. METHODS The sample included of 110 PWH on antiretroviral therapy. Fluid-attenuated inversion recovery (FLAIR) and T1-weighted anatomical MRI scans were collected, along with neuropsychological testing. FLAIR images were processed using the Lesion Segmentation Toolbox. A hierarchical regression model was run to investigate predictors of WMH burden [block 1: demographics; block 2: cerebrovascular disease (CVD) risk; block 3: lesion burden]. RESULTS The sample was 20% female and 79% African American with a mean age of 45.37. All participants had persistent HIV viral suppression, and the median CD4+ T-cell count was 750. Nearly a third (29%) currently used cocaine regularly, with an average of 23.75 (SD = 20.95) days in the past 90. In the hierarchical linear regression model, cocaine use was a significant predictor of WMH burden (β = .28). WMH burden was significantly correlated with poorer cognitive function (r = -0.27). Finally, higher WMH burden was significantly associated with increased serum concentrations of interferon-γ-inducible protein 10 (IP-10) but lower concentrations of myeloperoxidase (MPO); however, these markers did not differ by COC status. CONCLUSIONS WMH burden is associated with poorer cognitive performance in PWH. Cocaine use and CVD risk independently contribute to WMH, and addressing these conditions as part of HIV care may mitigate brain injury underlying neurocognitive impairment.
Collapse
Affiliation(s)
- Christina S. Meade
- Department of Psychiatry and Behavioral SciencesDuke University School of MedicineDurhamNorth Carolina27710USA
- Brain Imaging and Analysis CenterDuke University Medical CenterDurhamNorth Carolina27710USA
| | - Ryan P. Bell
- Department of Psychiatry and Behavioral SciencesDuke University School of MedicineDurhamNorth Carolina27710USA
| | - Sheri L. Towe
- Department of Psychiatry and Behavioral SciencesDuke University School of MedicineDurhamNorth Carolina27710USA
| | - Christopher D. Lascola
- Brain Imaging and Analysis CenterDuke University Medical CenterDurhamNorth Carolina27710USA
- Department of RadiologyDuke University School of MedicineDurhamNorth Carolina27710USA
| | - Kareem Al‐Khalil
- Department of Psychiatry and Behavioral SciencesDuke University School of MedicineDurhamNorth Carolina27710USA
| | - Matthew J. Gibson
- Department of Psychiatry and Behavioral SciencesDuke University School of MedicineDurhamNorth Carolina27710USA
| |
Collapse
|
33
|
Metwally SAH, Paruchuri SS, Yu L, Capuk O, Pennock N, Sun D, Song S. Pharmacological Inhibition of NHE1 Protein Increases White Matter Resilience and Neurofunctional Recovery after Ischemic Stroke. Int J Mol Sci 2023; 24:13289. [PMID: 37686096 PMCID: PMC10488118 DOI: 10.3390/ijms241713289] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
To date, recanalization interventions are the only available treatments for ischemic stroke patients; however, there are no effective therapies for reducing stroke-induced neuroinflammation. We recently reported that H+ extrusion protein Na+/H+ exchanger-1 (NHE1) plays an important role in stroke-induced inflammation and white matter injury. In this study, we tested the efficacy of two potent NHE1 inhibitors, HOE642 and Rimeporide, with a delayed administration regimen starting at 24 h post-stroke in adult C57BL/6J mice. Post-stroke HOE642 and Rimeporide treatments accelerated motor and cognitive function recovery without affecting the initial ischemic infarct, neuronal damage, or reactive astrogliosis. However, the delayed administration of NHE1 blockers after ischemic stroke significantly reduced microglial inflammatory activation while enhanced oligodendrogenesis and white matter myelination, with an increased proliferation and decreased apoptosis of the oligodendrocytes. Our findings suggest that NHE1 protein plays an important role in microglia-mediated inflammation and white matter damage. The pharmacological blockade of NHE1 protein activity reduced microglia inflammatory responses and enhanced oligodendrogenesis and white matter repair, leading to motor and cognitive function recovery after stroke. Our study reveals the potential of targeting NHE1 protein as a therapeutic strategy for ischemic stroke therapy.
Collapse
Affiliation(s)
- Shamseldin Ayman Hassan Metwally
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Satya Siri Paruchuri
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lauren Yu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Okan Capuk
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nicholas Pennock
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15213, USA
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (S.A.H.M.); (S.S.P.); (L.Y.); (O.C.); (N.P.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15213, USA
| |
Collapse
|
34
|
Ninomiya I, Koyama A, Otsu Y, Onodera O, Kanazawa M. Regeneration of the cerebral cortex by direct chemical reprogramming of macrophages into neuronal cells in acute ischemic stroke. Front Cell Neurosci 2023; 17:1225504. [PMID: 37636590 PMCID: PMC10457112 DOI: 10.3389/fncel.2023.1225504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Theoretically, direct chemical reprogramming of somatic cells into neurons in the infarct area represents a promising regenerative therapy for ischemic stroke. Previous studies have reported that human fibroblasts and astrocytes transdifferentiate into neuronal cells in the presence of small molecules without introducing ectopic transgenes. However, the optimal combination of small molecules for the transdifferentiation of macrophages into neurons has not yet been determined. The authors hypothesized that a combination of small molecules could induce the transdifferentiation of monocyte-derived macrophages into neurons and that the administration of this combination may be a regenerative therapy for ischemic stroke because monocytes and macrophages are directly involved in the ischemic area. Transcriptomes and morphologies of the cells were compared before and after stimulation using RNA sequencing and immunofluorescence staining. Microscopic analyses were also performed to identify cell markers and evaluate functional recovery by blinded examination following the administration of small molecules after ischemic stroke in CB-17 mice. In this study, an essential combination of six small molecules [CHIR99021, Dorsomorphin, Forskolin, isoxazole-9 (ISX-9), Y27632, and DB2313] that transdifferentiated monocyte-derived macrophages into neurons in vitro was identified. Moreover, administration of six small molecules after cerebral ischemia in model animals generated a new neuronal layer in the infarct cortex by converting macrophages into neuronal cells, ultimately improving neurological function. These results suggest that altering the transdifferentiation of monocyte-derived macrophages by the small molecules to adjust their adaptive response will facilitate the development of regenerative therapies for ischemic stroke.
Collapse
Affiliation(s)
- Itaru Ninomiya
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihide Koyama
- Department of Legal Medicine, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Yutaka Otsu
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
35
|
Li H, Xiao G, Tan X, Liu G, Xu Y, Gu S. Human umbilical cord blood mononuclear cells ameliorate ischemic brain injury via promoting microglia/macrophages M2 polarization in MCAO Rats. Exp Brain Res 2023; 241:1585-1598. [PMID: 37142782 DOI: 10.1007/s00221-023-06600-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023]
Abstract
Cerebral infarction is one of the most prevalent cerebrovascular disorders. Microglia and infiltrating macrophages play a key role in regulating the inflammatory response after ischemic stroke. Regulation of microglia/macrophages polarization contributes to the recovery of neurological function in cerebral infarction. In recent decades, human umbilical cord blood mononuclear cells (hUCBMNCs) have been considered a potential therapeutic alternative. However, the mechanism of action is yet unclear. Our study aimed to explore whether hUCBMNCs treatment for cerebral infarction is via regulation of microglia/macrophages polarization. Adult male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) and were treated by intravenous routine with or without hUCBMNCs at 24 h following MCAO. We evaluated the therapeutic effects of hUCBMNCs on cerebral infarction by measuring animal behavior and infarct volume, and further explored the possible mechanisms of hUCBMNCs for cerebral infarction by measuring inflammatory factors and microglia/macrophages markers using Elisa and immunofluorescence staining, respectively. We found that administration with hUCBMNCs improved behavioral functions and reduced infarct volume. Rats treated with hUCBMNCs showed a significant reduction in the level of IL-6, and TNF-α and increased the level of IL-4 and IL-10 compared to those treated without hUCBMNCs. Furthermore, hUCBMNCs inhibited M1 polarization and promoted M2 polarization of microglia/macrophages after MCAO. We conclude that hUCBMNCs could ameliorate cerebral brain injury by promoting microglia/macrophages M2 polarization in MCAO Rats. This experiment provides evidence that hUCBMNCs represent a promising therapeutic option for ischemic stroke.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Gai Xiao
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiao Tan
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Guojun Liu
- Shandong Cord Blood Bank, Jinan, Shangdong, China
| | - Yangzhou Xu
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shaojuan Gu
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
36
|
Kamal FZ, Lefter R, Jaber H, Balmus IM, Ciobica A, Iordache AC. The Role of Potential Oxidative Biomarkers in the Prognosis of Acute Ischemic Stroke and the Exploration of Antioxidants as Possible Preventive and Treatment Options. Int J Mol Sci 2023; 24:ijms24076389. [PMID: 37047362 PMCID: PMC10094154 DOI: 10.3390/ijms24076389] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Ischemic strokes occur when the blood supply to a part of the brain is interrupted or reduced due to arterial blockage, and it often leads to damage to brain cells or death. According to a myriad of experimental studies, oxidative stress is an important pathophysiological mechanism of ischemic stroke. In this narrative review, we aimed to identify how the alterations of oxidative stress biomarkers could suggest a severity-reflecting diagnosis of ischemic stroke and how these interactions may provide new molecular targets for neuroprotective therapies. We performed an eligibility criteria-based search on three main scientific databases. We found that patients with acute ischemic stroke are characterized by increased oxidative stress markers levels, such as the total antioxidant capacity, F2-isoprostanes, hydroxynonenal, total and perchloric acid oxygen radical absorbance capacity (ORACTOT and ORACPCA), malondialdehyde (MDA), myeloperoxidase, and urinary 8-oxo-7,8-dihydro-2′-deoxyguanosine. Thus, acute ischemic stroke is causing significant oxidative stress and associated molecular and cellular damage. The assessment of these molecular markers could be useful in diagnosing ischemic stroke, finding its causes, predicting its severity and outcomes, reducing its impact on the cellular structures of the brain, and guiding preventive treatment towards antioxidant-based therapy as novel therapeutic alternatives.
Collapse
|
37
|
Xie M, Hao Y, Feng L, Wang T, Yao M, Li H, Ma D, Feng J. Neutrophil Heterogeneity and its Roles in the Inflammatory Network after Ischemic Stroke. Curr Neuropharmacol 2023; 21:621-650. [PMID: 35794770 PMCID: PMC10207908 DOI: 10.2174/1570159x20666220706115957] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
As the first peripheral immune cells to enter the brain after ischemic stroke, neutrophils are important participants in stroke-related neuroinflammation. Neutrophils are quickly mobilized from the periphery in response to a stroke episode and cross the blood-brain barrier to reach the ischemic brain parenchyma. This process involves the mobilization and activation of neutrophils from peripheral immune organs (including the bone marrow and spleen), their chemotaxis in the peripheral blood, and their infiltration into the brain parenchyma (including disruption of the blood-brain barrier, inflammatory effects on brain tissue, and interactions with other immune cell types). In the past, it was believed that neutrophils aggravated brain injuries through the massive release of proteases, reactive oxygen species, pro-inflammatory factors, and extracellular structures known as neutrophil extracellular traps (NETs). With the failure of early clinical trials targeting neutrophils and uncovering their underlying heterogeneity, our view of their role in ischemic stroke has become more complex and multifaceted. As neutrophils can be divided into N1 and N2 phenotypes in tumors, neutrophils have also been found to have similar phenotypes after ischemic stroke, and play different roles in the development and prognosis of ischemic stroke. N1 neutrophils are dominant during the acute phase of stroke (within three days) and are responsible for the damage to neural structures via the aforementioned mechanisms. However, the proportion of N2 neutrophils gradually increases in later phases, and this has a beneficial effect through the release of anti-inflammatory factors and other neuroprotective mediators. Moreover, the N1 and N2 phenotypes are highly plastic and can be transformed into each other under certain conditions. The pronounced differences in their function and their high degree of plasticity make these neutrophil subpopulations promising targets for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Meizhen Xie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Liangshu Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Tian Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Mengyue Yao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| |
Collapse
|
38
|
Wang Y, Jia Y, Xu Q, Wang R, Sun L, Guo D, Shi M, Yang P, Wang Y, Liu F, Zhang Y, Zhu Z. Association between myeloperoxidase and the risks of ischemic stroke, heart failure, and atrial fibrillation: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2023; 33:210-218. [PMID: 36411224 DOI: 10.1016/j.numecd.2022.09.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/13/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIMS The causality between myeloperoxidase (MPO) and cardiovascular disease still remains unclear. We performed a two-sample Mendelian randomization (MR) study to estimate the potential causal effect of MPO on the risks of ischemic stroke, ischemic stroke subtypes, heart failure (HF), and atrial fibrillation (AF). METHODS AND RESULTS Seventeen independent single-nucleotide polymorphisms associated with MPO levels were identified as instrumental variables from a European-descent genome-wide association study. Summary-level data on ischemic stroke originated from the Multiancestry Genome-wide Association Study of Stroke Consortium with 440 328 European individuals. We used the inverse-variance weighted method to assess the potential causality of plasma MPO with ischemic stroke and its subtypes in the main analysis. Genetically determined higher plasma MPO concentration was significantly associated with increased risks of ischemic stroke (odds ratio [OR] per standard deviation [SD] increase, 1.05; 95% confidence interval [CI], 1.02-1.09; P = 0.002) and cardioembolic stroke (CES) (OR per SD increase, 1.10; 95% CI, 1.03-1.18; P = 0.005), but was not associated with risks of large artery stroke or small vessel stroke. In the secondary analysis, MPO was associated with a high risk of HF (OR per SD increase, 1.05; 95% CI, 1.02-1.07; P = 0.001) and AF (OR per SD increase, 1.04; 95% CI, 1.01-1.07; P = 0.004). MR-Egger regression showed no directional pleiotropy for all associations, and the sensitivity analyses further confirmed these findings. CONCLUSION High plasma MPO levels were potentially associated with increased risks of ischemic stroke, CES, HF, and AF, suggesting that MPO plays an important role in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Yinan Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qingyun Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ruirui Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Daoxia Guo
- School of Nursing, Suzhou Medical College of Soochow University, Suzhou, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yu Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Fanghua Liu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
39
|
Tao H, Dong L, Li L. N6-methyladenosine modulation classes and immune microenvironment regulation in ischemic stroke. Front Mol Neurosci 2022; 15:1013076. [PMID: 36762188 PMCID: PMC9907088 DOI: 10.3389/fnmol.2022.1013076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/07/2022] [Indexed: 12/25/2022] Open
Abstract
N6-methyladenosine (m6A) modifications play an important role in the differentiation and regulation of immune cells. However, research on m6A in ischemic stroke (IS) is still in its infancy, and their role of the immune microenvironment remains unknown. In this study, we systematically assessed the modification classes of m6A regulators in IS based on the GEO database (GSE16561 and GSE22255). We found that in IS patients, IGF2BP2, IGF2BP1, and YTHDF2 expression was significantly upregulated, and ELAVL1, LRPPRC, METTL3, ALKBH5, CBLL1, and METTL14 expression was significantly downregulated. Seven IS-related genes (ELAVL1, IGF2BP2, LRPPRC, YTHDF2, ALKBH5, METTL14, and YTHDC1) were finally screened by logistic and least absolute shrinkage and selection operator (LASSO) regressions, and the AUC of the riskScore was 0.942, which was a good classification. For immune infiltration, there were highly significant differences in memory B cells, CD8 T cells, monocytes, activated dendritic cells, and mast cells between IS and normal samples. The IS samples were grouped into three classes by consistent clustering, and 15 m6A genes were differentially expressed in the different classes. Multiple infiltrating immune cells, immune-associated genes, and HLA-associated genes differed significantly across m6A modification classes, indicating the diversity and complexity of m6A modifications in the immune microenvironment of IS. Finally, 487 genes associated with the m6A modification class were identified, and 227 potential drugs were found. Our findings demonstrated that m6A modification plays a crucial role in the immune regulation of IS.
Collapse
Affiliation(s)
- Hongmiao Tao
- Medical College, Jinhua Polytechnic, Jinhua, China,*Correspondence: Hongmiao Tao,
| | - Lihua Dong
- Medical College, Jinhua Polytechnic, Jinhua, China
| | - Lin Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
40
|
Towards a Whole Sample Imaging Approach Using Diffusion Tensor Imaging to Examine the Foreign Body Response to Explanted Medical Devices. Polymers (Basel) 2022; 14:polym14224819. [PMID: 36432947 PMCID: PMC9698821 DOI: 10.3390/polym14224819] [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: 08/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
Analysing the composition and organisation of the fibrous capsule formed as a result of the Foreign Body Response (FBR) to medical devices, is imperative for medical device improvement and biocompatibility. Typically, analysis is performed using histological techniques which often involve random sampling strategies. This method is excellent for acquiring representative values but can miss the unique spatial distribution of features in 3D, especially when analysing devices used in large animal studies. To overcome this limitation, we demonstrate a non-destructive method for high-resolution large sample imaging of the fibrous capsule surrounding human-sized implanted devices using diffusion tensor imaging (DTI). In this study we analyse the fibrous capsule surrounding two unique macroencapsulation devices that have been implanted in a porcine model for 21 days. DTI is used for 3D visualisation of the microstructural organisation and validated using the standard means of fibrous capsule investigation; histological analysis and qualitative micro computed tomography (microCT) and scanning electron microscopy (SEM) imaging. DTI demonstrated the ability to distinguish microstructural differences in the fibrous capsules surrounding two macroencapsulation devices made from different materials and with different surface topographies. DTI-derived metrics yielded insight into the microstructural organisation of both capsules which was corroborated by microCT, SEM and histology. The non-invasive characterisation of the integration of implants in the body has the potential to positively influence analysis methods in pre-clinical studies and accelerate the clinical translation of novel implantable devices.
Collapse
|
41
|
Koehn LM, Nguyen K, Chen X, Santoso A, Tucker R, Lim YP, Stonestreet BS. Effects of Three Different Doses of Inter-Alpha Inhibitor Proteins on Severe Hypoxia-Ischemia-Related Brain Injury in Neonatal Rats. Int J Mol Sci 2022; 23:13473. [PMID: 36362257 PMCID: PMC9655902 DOI: 10.3390/ijms232113473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2024] Open
Abstract
Hypoxia-ischemia (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. We have previously shown that human plasma-derived inter-alpha inhibitor proteins (hIAIPs) attenuate HI-related brain injury in neonatal rats. The optimal dose of hIAIPs for their neuroprotective effects and improvement in behavioral outcomes remains to be determined. We examined the efficacy of 30, 60, or 90 mg/kg of hIAIPs administered to neonatal rats after exposure to HI for 2 h. Postnatal day 7 (P7) Wistar rats were exposed to either sham-surgery or unilateral HI (right carotid artery ligation, 2 h of 8% O2) brain injury. A placebo, 30, 60, or 90 mg/kg of hIAIPs were injected intraperitoneally at 0, 24 and 48 h after HI (n = 9-10/sex). We carried out the following behavioral analyses: P8 (righting reflex), P9 (negative geotaxis) and P10 (open-field task). Rats were humanely killed on P10 and their brains were stained with cresyl violet. Male extension/contraction responses and female righting reflex times were higher in the HI placebo groups than the sham groups. Female open-field exploration was lower in the HI placebo group than the sham group. hIAIPs attenuated these behavioral deficits. However, the magnitude of the responses did not vary by hIAIP dose. hIAIPs reduced male brain infarct volumes in a manner that correlated with improved behavioral outcomes. Increasing the hIAIP dose from 30 to 90 mg/kg did not further accentuate the hIAIP-related decreases in infarct volumes. We conclude that larger doses of hIAIPs did not provide additional benefits over the 30 mg/kg dose for behavior tasks or reductions in infarct volumes in neonatal rats after exposure to severe HI.
Collapse
Affiliation(s)
- Liam M. Koehn
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Kevin Nguyen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | | | - Richard Tucker
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
42
|
Li Y, Xia Q, Zhu C, Cao W, Xia Z, Liu X, Xiao B, Chen K, Liu Y, Zhong L, Tan B, Lei J, Zhu J. An activatable Mn(II) MRI probe for detecting peroxidase activity in vitro and in vivo. J Inorg Biochem 2022; 236:111979. [PMID: 36087435 DOI: 10.1016/j.jinorgbio.2022.111979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 12/15/2022]
Abstract
Myeloperoxidase (MPO), a hallmark of the function and activation of innate immune cells, can act as a 'double-edged sword', contributing to clear infection as well as causing tissue oxidizing damage in various inflammatory diseases. In this study, an activatable Mn(II) chelate-based magnetic resonance imaging (MRI) contrast agent (CA), Mn-TyEDTA (TyEDTA = tyrosine derived ethylenediaminetetraacetic acid) structurally featuring a phenol group as the electron-donor, was developed to sense the activity of peroxidase in vitro and in vivo. Mn-TyEDTA demonstrated a peroxidase activity-dependent relaxivity in the presence of horseradish peroxidase (HRP)/H2O2 with more than a 2.6-fold increase in water proton relaxivity produced (HRP, 500 U; H2O2, 4.5 eq). A mechanism of peroxidase-mediated Mn(II) monomer radical polymerization was confirmed with those oligomers of Mn-TyEDTA such as dimer, trimer and tetramer were found in the LC-MS study. Dynamic MR imaging of normal mice revealed rapid blood clearance and mixed renal and hepatobiliary elimination of Mn-TyEDTA. Furthermore, compared to liver-specific and non-specific extracellular contrast agents (Mn-BnO-TyEDTA (BnO-TyEDTA = benzyl tyrosine-derived ethylenediaminetetraacetic acid) and Gd-DTPA (DTPA = diethylene triamine penta-acetic acid)), MRI on a monosodium urate (MSU) crystal-induced acute mice model of arthritis showed that inflamed tissues could be selectively enhanced by Mn-TyEDTA, suggesting that this peroxidase-activatable Mn(II) MRI probe could potentially be used for noninvasive detection of MPO activity in vivo.
Collapse
Affiliation(s)
- Yunhe Li
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China
| | - Qian Xia
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Chunrong Zhu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Weidong Cao
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China
| | - Zhiyang Xia
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Xinxin Liu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Bin Xiao
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Keyu Chen
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China
| | - Yun Liu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Lei Zhong
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Bangxian Tan
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China
| | - Jun Lei
- School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China.
| | - Jiang Zhu
- Sichuan Key Laboratory of Medical Imaging, Department of Oncology, and Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Maoyuan Road 1, Nanchong City, Sichuan 637000, China; School of Pharmacy, North Sichuan Medical College, Fujiang Road 234, Nanchong City, Sichuan 637000, China.
| |
Collapse
|
43
|
Sternak M, Glasnović A, Josić P, Romić D, Gajović S. The effects of splenectomy in murine models of ischemic stroke: a systematic review and meta-analysis. J Neuroinflammation 2022; 19:233. [PMID: 36151564 PMCID: PMC9508771 DOI: 10.1186/s12974-022-02593-w] [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: 01/26/2022] [Accepted: 09/13/2022] [Indexed: 12/09/2022] Open
Abstract
Background The spleen, a substantial reservoir of non-differentiated monocytes, may play a crucial role in the pathophysiology of post-ischemic inflammation and influence outcomes after ischemic stroke.
Aim of the study To analyze splenectomy as a preclinical intervention in murine models of ischemic stroke. Methods Following systematic searches of PubMed, Scopus and Web of Science, a qualitative synthesis of study characteristics was performed, and the effect of splenectomy estimated by a three-level random-effects meta-analysis of infarct volumes and a conventional two-level random-effects meta-analysis of neurological deficit scores. Results Database searches identified a total of 14 studies, 13 of which were used for meta-analysis. The ischemic lesion volumes were reduced in splenectomized animals compared to the control groups (difference in standardized mean differences: − 1.42; 95% CI [− 1.98, − 0.85]; 95% PI [− 2.03, − 0.80]; I2(2) = 19.04%; 95% CI [0.00%, 65.49%]; I2(3) = 47.24%; 95% CI [0.00%, 85.23%]) and neurological deficit scores were improved (− 1.20; 95% CI [− 2.20, − 0.20]; 95% PI [− 4.58, 2.18]; I2 = 77.5%; 95% CI [50.0%, 89.9%]). A subgroup analysis for infarct volumes showed that splenectomy performed prior to ischemia achieved a higher reduction of the ischemic lesion than when splenectomy was performed immediately prior or after stroke. Although the overall effect size of splenectomy could be classified as large, there was a significant presence of risks of bias, study heterogeneity, and a potential presence of publication bias. Conclusion Despite limitations related to heterogeneity, risks of bias, and potential publication bias, this meta-analysis points to the spleen and its functional cell populations as promising targets for the therapeutic modulation of post-stroke inflammation. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02593-w.
Collapse
Affiliation(s)
- Marko Sternak
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Anton Glasnović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Paula Josić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Dominik Romić
- Department of Neurosurgery, University of Zagreb School of Medicine, University Hospital Dubrava, Zagreb, Croatia
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia.
| |
Collapse
|
44
|
Santos-Lima B, Pietronigro EC, Terrabuio E, Zenaro E, Constantin G. The role of neutrophils in the dysfunction of central nervous system barriers. Front Aging Neurosci 2022; 14:965169. [PMID: 36034148 PMCID: PMC9404376 DOI: 10.3389/fnagi.2022.965169] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Leukocyte migration into the central nervous system (CNS) represents a central process in the development of neurological diseases with a detrimental inflammatory component. Infiltrating neutrophils have been detected inside the brain of patients with several neuroinflammatory disorders, including stroke, multiple sclerosis and Alzheimer’s disease. During inflammatory responses, these highly reactive innate immune cells can rapidly extravasate and release a plethora of pro-inflammatory and cytotoxic factors, potentially inducing significant collateral tissue damage. Indeed, several studies have shown that neutrophils promote blood-brain barrier damage and increased vascular permeability during neuroinflammatory diseases. Recent studies have shown that neutrophils migrate into the meninges and choroid plexus, suggesting these cells can also damage the blood-cerebrospinal fluid barrier (BCSFB). In this review, we discuss the emerging role of neutrophils in the dysfunction of brain barriers across different neuroinflammatory conditions and describe the molecular basis and cellular interplays involved in neutrophil-mediated injury of the CNS borders.
Collapse
|
45
|
Wang X, Zhang L, Li P, Zheng Y, Yang Y, Ji S. Apelin/APJ system in inflammation. Int Immunopharmacol 2022; 109:108822. [DOI: 10.1016/j.intimp.2022.108822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 12/18/2022]
|
46
|
Wicks EE, Ran KR, Kim JE, Xu R, Lee RP, Jackson CM. The Translational Potential of Microglia and Monocyte-Derived Macrophages in Ischemic Stroke. Front Immunol 2022; 13:897022. [PMID: 35795678 PMCID: PMC9251541 DOI: 10.3389/fimmu.2022.897022] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune response to ischemic stroke is an area of study that is at the forefront of stroke research and presents promising new avenues for treatment development. Upon cerebral vessel occlusion, the innate immune system is activated by danger-associated molecular signals from stressed and dying neurons. Microglia, an immune cell population within the central nervous system which phagocytose cell debris and modulate the immune response via cytokine signaling, are the first cell population to become activated. Soon after, monocytes arrive from the peripheral immune system, differentiate into macrophages, and further aid in the immune response. Upon activation, both microglia and monocyte-derived macrophages are capable of polarizing into phenotypes which can either promote or attenuate the inflammatory response. Phenotypes which promote the inflammatory response are hypothesized to increase neuronal damage and impair recovery of neuronal function during the later phases of ischemic stroke. Therefore, modulating neuroimmune cells to adopt an anti-inflammatory response post ischemic stroke is an area of current research interest and potential treatment development. In this review, we outline the biology of microglia and monocyte-derived macrophages, further explain their roles in the acute, subacute, and chronic stages of ischemic stroke, and highlight current treatment development efforts which target these cells in the context of ischemic stroke.
Collapse
|
47
|
Zhao Y, Gan L, Ren L, Lin Y, Ma C, Lin X. Factors influencing the blood-brain barrier permeability. Brain Res 2022; 1788:147937. [PMID: 35568085 DOI: 10.1016/j.brainres.2022.147937] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic structure that protects the brain from harmful blood-borne, endogenous and exogenous substances and maintains the homeostatic microenvironment. All constituent cell types play indispensable roles in the BBB's integrity, and other structural BBB components, such as tight junction proteins, adherens junctions, and junctional proteins, can control the barrier permeability. Regarding the need to exchange nutrients and toxic materials, solute carriers, ATP-binding case families, and ion transporter, as well as transcytosis regulate the influx and efflux transport, while the difference in localisation and expression can contribute to functional differences in transport properties. Numerous chemical mediators and other factors such as non-physicochemical factors have been identified to alter BBB permeability by mediating the structural components and barrier function, because of the close relationship with inflammation. In this review, we highlight recently gained mechanistic insights into the maintenance and disruption of the BBB. A better understanding of the factors influencing BBB permeability could contribute to supporting promising potential therapeutic targets for protecting the BBB and the delivery of central nervous system drugs via BBB permeability interventions under pathological conditions.
Collapse
Affiliation(s)
- Yibin Zhao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Gan
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Ren
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yubo Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Congcong Ma
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianming Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
48
|
Candelario-Jalil E, Dijkhuizen RM, Magnus T. Neuroinflammation, Stroke, Blood-Brain Barrier Dysfunction, and Imaging Modalities. Stroke 2022; 53:1473-1486. [PMID: 35387495 PMCID: PMC9038693 DOI: 10.1161/strokeaha.122.036946] [Citation(s) in RCA: 390] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Maintaining blood-brain barrier (BBB) integrity is crucial for the homeostasis of the central nervous system. Structurally comprising the BBB, brain endothelial cells interact with pericytes, astrocytes, neurons, microglia, and perivascular macrophages in the neurovascular unit. Brain ischemia unleashes a profound neuroinflammatory response to remove the damaged tissue and prepare the brain for repair. However, the intense neuroinflammation occurring during the acute phase of stroke is associated with BBB breakdown, neuronal injury, and worse neurological outcomes. Here, we critically discuss the role of neuroinflammation in ischemic stroke pathology, focusing on the BBB and the interactions between central nervous system and peripheral immune responses. We highlight inflammation-driven injury mechanisms in stroke, including oxidative stress, increased MMP (matrix metalloproteinase) production, microglial activation, and infiltration of peripheral immune cells into the ischemic tissue. We provide an updated overview of imaging techniques for in vivo detection of BBB permeability, leukocyte infiltration, microglial activation, and upregulation of cell adhesion molecules following ischemic brain injury. We discuss the possibility of clinical implementation of imaging modalities to assess stroke-associated neuroinflammation with the potential to provide image-guided diagnosis and treatment. We summarize the results from several clinical studies evaluating the efficacy of anti-inflammatory interventions in stroke. Although convincing preclinical evidence suggests that neuroinflammation is a promising target for ischemic stroke, thus far, translating these results into the clinical setting has proved difficult. Due to the dual role of inflammation in the progression of ischemic damage, more research is needed to mechanistically understand when the neuroinflammatory response begins the transition from injury to repair. This could have important implications for ischemic stroke treatment by informing time- and context-specific therapeutic interventions.
Collapse
Affiliation(s)
- Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville (E.C-J)
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, the Netherlands (R.M.D.)
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Germany (T.M.)
| |
Collapse
|
49
|
Re-directing nanomedicines to the spleen: A potential technology for peripheral immunomodulation. J Control Release 2022; 350:60-79. [DOI: 10.1016/j.jconrel.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
|
50
|
Smyth LCD, Murray HC, Hill M, van Leeuwen E, Highet B, Magon NJ, Osanlouy M, Mathiesen SN, Mockett B, Singh-Bains MK, Morris VK, Clarkson AN, Curtis MA, Abraham WC, Hughes SM, Faull RLM, Kettle AJ, Dragunow M, Hampton MB. Neutrophil-vascular interactions drive myeloperoxidase accumulation in the brain in Alzheimer's disease. Acta Neuropathol Commun 2022; 10:38. [PMID: 35331340 PMCID: PMC8944147 DOI: 10.1186/s40478-022-01347-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/11/2022] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Neutrophil accumulation is a well-established feature of Alzheimer's disease (AD) and has been linked to cognitive impairment by modulating disease-relevant neuroinflammatory and vascular pathways. Neutrophils express high levels of the oxidant-generating enzyme myeloperoxidase (MPO), however there has been controversy regarding the cellular source and localisation of MPO in the AD brain. MATERIALS AND METHODS We used immunostaining and immunoassays to quantify the accumulation of neutrophils in human AD tissue microarrays and in the brains of APP/PS1 mice. We also used multiplexed immunolabelling to define the presence of NETs in AD. RESULTS There was an increase in neutrophils in AD brains as well as in the murine APP/PS1 model of AD. Indeed, MPO expression was almost exclusively confined to S100A8-positive neutrophils in both human AD and murine APP/PS1 brains. The vascular localisation of neutrophils in both human AD and mouse models of AD was striking and driven by enhanced neutrophil adhesion to small vessels. We also observed rare infiltrating neutrophils and deposits of MPO around plaques. Citrullinated histone H3, a marker of neutrophil extracellular traps (NETs), was also detected in human AD cases at these sites, indicating the presence of extracellular MPO in the vasculature. Finally, there was a reduction in the endothelial glycocalyx in AD that may be responsible for non-productive neutrophil adhesion to the vasculature. CONCLUSION Our report indicates that vascular changes may drive neutrophil adhesion and NETosis, and that neutrophil-derived MPO may lead to vascular oxidative stress and be a relevant therapeutic target in AD.
Collapse
Affiliation(s)
- Leon C. D. Smyth
- Centre for Free Radical Research, University of Otago, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140 New Zealand
- Department of Pathology and Immunology, Center for Brain Immunology and Glia, Washington University in St. Louis, Campus, Box 8118, St. Louis, MO USA
| | - Helen C. Murray
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy With Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Madison Hill
- Centre for Free Radical Research, University of Otago, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140 New Zealand
| | - Eve van Leeuwen
- Centre for Free Radical Research, University of Otago, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140 New Zealand
| | - Blake Highet
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy With Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Nicholas J. Magon
- Centre for Free Radical Research, University of Otago, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140 New Zealand
| | - Mahyar Osanlouy
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Sophie N. Mathiesen
- Department of Psychology, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Bruce Mockett
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Malvindar K. Singh-Bains
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy With Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Vanessa K. Morris
- School of Biological Science, University of Canterbury, Canterbury, New Zealand
| | | | - Maurice A. Curtis
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy With Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | - Richard L. M. Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy With Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Anthony J. Kettle
- Centre for Free Radical Research, University of Otago, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140 New Zealand
| | - Mike Dragunow
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Mark B. Hampton
- Centre for Free Radical Research, University of Otago, Christchurch, New Zealand
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140 New Zealand
| |
Collapse
|