1
|
Saiprayong K, Chupradit K, Sasithong P, Suwanpitak S, Muneekaew S, Thongsin N, Srisantitham J, Wattanapanitch M. Development of 2LTRZFP-expressing induced pluripotent stem cells as a potential anti-HIV-1 gene therapy against viral integration. J Leukoc Biol 2025; 117:qiaf018. [PMID: 39946247 DOI: 10.1093/jleuko/qiaf018] [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: 06/28/2024] [Revised: 11/29/2024] [Accepted: 02/12/2025] [Indexed: 04/26/2025] Open
Abstract
Highly active antiretroviral drug is the standard treatment for HIV-1 infection to suppress the viral load. However, this treatment does not completely eradicate the virus; it simply decreases the viral load to undetectable levels. The development of a novel therapy to cure the disease is essential. Previously, we developed an engineered zinc finger protein (ZFP) that specifically binds to the 2-LTR-circle junction (2LTRZFP), the target site for viral integrase, preventing HIV-1 integration in human CD34+ hematopoietic stem/progenitor cells (HSPCs) and macrophages. Although the transduction efficiency of 2LTRZFP was ∼50%, purifying and expanding the 2LTRZFP-expressing HSPCs proved difficult. In addition, the batch-to-batch variability in transduction efficiency could have a major impact on the therapeutic efficacy. In this study, we introduced the 2LTRZFP into human induced pluripotent stem cells (iPSCs) followed by clonal isolation and functional validation of the 2LTRZFP. Upon the HIV-1 challenge, the 2LTRZFP protein was found to inhibit the viral integration in iPSCs, iPSC-derived HSPCs, and macrophages. The engineered iPSC clone could be differentiated into functional macrophages, as evidenced by M1 and M2 polarization, and phagocytosis. Our finding revealed that the 2LTRZFP did not perturb the macrophage differentiation process. Therefore, the 2LTRZFP-expressing iPSCs could provide an unlimited supply of HIV-1-resistant HSPCs for transplantation, potentially leading to HIV-1-resistant blood cells. The knowledge obtained from this study will provide a cornerstone for HIV-1 gene therapy using HSPC transplantation as a sustainable HIV-1 treatment in the future.
Collapse
Affiliation(s)
- Kritayaporn Saiprayong
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Koollawat Chupradit
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Pasut Sasithong
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Biomedical Sciences Graduate Program, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Siriwal Suwanpitak
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Saitong Muneekaew
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Nontaphat Thongsin
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Jakkrapatra Srisantitham
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Methichit Wattanapanitch
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| |
Collapse
|
2
|
Nixon RA, Rubinsztein DC. Mechanisms of autophagy-lysosome dysfunction in neurodegenerative diseases. Nat Rev Mol Cell Biol 2024; 25:926-946. [PMID: 39107446 DOI: 10.1038/s41580-024-00757-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/15/2024]
Abstract
Autophagy is a lysosome-based degradative process used to recycle obsolete cellular constituents and eliminate damaged organelles and aggregate-prone proteins. Their postmitotic nature and extremely polarized morphologies make neurons particularly vulnerable to disruptions caused by autophagy-lysosomal defects, especially as the brain ages. Consequently, mutations in genes regulating autophagy and lysosomal functions cause a wide range of neurodegenerative diseases. Here, we review the role of autophagy and lysosomes in neurodegenerative diseases such as Alzheimer disease, Parkinson disease and frontotemporal dementia. We also consider the strong impact of cellular ageing on lysosomes and autophagy as a tipping point for the late-age emergence of related neurodegenerative disorders. Many of these diseases have primary defects in autophagy, for example affecting autophagosome formation, and in lysosomal functions, especially pH regulation and calcium homeostasis. We have aimed to provide an integrative framework for understanding the central importance of autophagic-lysosomal function in neuronal health and disease.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| |
Collapse
|
3
|
Jing K, Gu R, Chen F, Wan J, Sun Y, Guo P, Chen F, Feng J, Guo J, Liu X. Orosomucoid 2 is an endogenous regulator of neuronal mitochondrial biogenesis and promotes functional recovery post-stroke. Pharmacol Res 2024; 209:107422. [PMID: 39293585 DOI: 10.1016/j.phrs.2024.107422] [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: 03/10/2024] [Revised: 09/01/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024]
Abstract
Development of functional recovery therapies is critical to reduce the global impact of stroke as the leading cause of long-term disability. Our previous studies found that acute-phase protein orosomucoid (ORM) could provide an up to 6 h therapeutic time window to reduce infarct volume in acute ischemic stroke by improving endothelial function. However, its role in neurons and functional recovery post-stroke remains largely unknown. Here, we showed that exogenous ORM administration with initial injection at 0.5 h (early) or 12 h (delayed) post-MCAO daily for consecutive 7 days significantly decreased infarct area, improved motor and cognitive functional recovery, and promoted mitochondrial biogenesis after MCAO. While neuron-specific knockout of ORM2, a dominant subtype of ORM in the brain, produced opposite effects which could be rescued by exogenous ORM. In vitro, exogenous ORM protected SH-SY5Y cells from OGD-induced damage and promoted mitochondrial biogenesis, while endogenous ORM2 deficiency worsened these processes. Mechanistically, inactivation of CCR5 or AMPK eliminated the protective effects of ORM on neuronal damage and mitochondrial biogenesis. Taken together, our findings demonstrate that ORM, mainly ORM2, is an endogenous regulator of neuronal mitochondrial biogenesis by activating CCR5/AMPK signaling pathway, and might act as a potential therapeutic target for the functional recovery post-stroke.
Collapse
Affiliation(s)
- Kai Jing
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Ruinan Gu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Feng Chen
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Jingjing Wan
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Yang Sun
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Pengyue Guo
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Fei Chen
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Jiayi Feng
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China
| | - Jinmin Guo
- Department of Clinical Pharmacy, 960th Hospital of Joint Logistic Support Force, Jinan, Shandong, China.
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200082, China.
| |
Collapse
|
4
|
Li Y, Wan TT, Li JX, Xiao X, Liu L, Li HH, Guo SB. ACE2 Rescues Sepsis-Associated Encephalopathy by Reducing Inflammation, Oxidative Stress, and Neuronal Apoptosis via the Nrf2/Sestrin2 Signaling Pathway. Mol Neurobiol 2024; 61:8640-8655. [PMID: 38532242 DOI: 10.1007/s12035-024-04063-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Neuroinflammation and oxidative stress contribute to the progression of sepsis-associated encephalopathy (SAE). Angiotensin-converting enzyme 2 (ACE2) is considered to be a neuroprotective factor due to its anti-inflammatory and antioxidant properties. However, the role of ACE2 on myeloid cells in regulating SAE and the underlying mechanism warrants further exploration. SAE was induced in ACE2 transgenic (TG), knockout (KO), and bone marrow (BM) chimeric mice by cecal ligation and puncture (CLP). The expression levels of apoptosis-, oxidation- and neuroinflammation-associated mediators and morphological changes were monitored by quantitative real-time PCR analyses and histological examinations in the cortex of septic mice. The contents of angiotensin (Ang) II and Ang-(1-7) along with the activity of ACE2 were examined with commercial kits. The expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and Sestrin2 was detected by immunoblotting analysis. Our results indicated that the expression of cortical ACE2 was significantly reduced in the early phase of CLP-induced sepsis. Moreover, ACE2 overexpression in TG mice conferred neuroprotection against sepsis, as evidenced by alleviated neuronal apoptosis, oxidative stress, and proinflammatory M1-like microglial polarization, accompanied by upregulation of the Ang-(1-7), Nrf2, and Sestrin2 protein levels. Conversely, ACE2 deficiency in KO mice exacerbated SAE. The neuroprotective effects of ACE2 were further confirmed in wild-type mice transplanted with ACE2-TG and KO BM cells. Therefore, our data suggest that myeloid ACE2 exerts a protective role in the pathogenesis of SAE, potentially by activating Ang-(1-7)-Nrf2/sestrin2 signaling pathway, and highlight that upregulating ACE2 expression and activity may represent a promising approach for the treatment of SAE in patients with sepsis.
Collapse
Affiliation(s)
- Ya Li
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Tian-Tian Wan
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jia-Xin Li
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xue Xiao
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lei Liu
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hui-Hua Li
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Shu-Bin Guo
- Emergency Medicine Clinical Research Center, Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
5
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
6
|
Tournier BB, Sorce S, Marteyn A, Ghidoni R, Benussi L, Binetti G, Herrmann FR, Krause K, Zekry D. CCR5 deficiency: Decreased neuronal resilience to oxidative stress and increased risk of vascular dementia. Alzheimers Dement 2024; 20:124-135. [PMID: 37489764 PMCID: PMC10917026 DOI: 10.1002/alz.13392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
INTRODUCTION As the chemokine receptor5 (CCR5) may play a role in ischemia, we studied the links between CCR5 deficiency, the sensitivity of neurons to oxidative stress, and the development of dementia. METHODS Logistic regression models with CCR5/apolipoprotein E (ApoE) polymorphisms were applied on a sample of 205 cognitively normal individuals and 189 dementia patients from Geneva. The impact of oxidative stress on Ccr5 expression and cell death was assessed in mice neurons. RESULTS CCR5-Δ32 allele synergized with ApoEε4 as risk factor for dementia and specifically for dementia with a vascular component. We confirmed these results in an independent cohort from Italy (157 cognitively normal and 620 dementia). Carriers of the ApoEε4/CCR5-Δ32 genotype aged ≥80 years have an 11-fold greater risk of vascular-and-mixed dementia. Oxidative stress-induced cell death in Ccr5-/- mice neurons. DISCUSSION We propose the vulnerability of CCR5-deficient neurons in response to oxidative stress as possible mechanisms contributing to dementia.
Collapse
Affiliation(s)
- Benjamin B. Tournier
- Department of PsychiatryGeneva University Hospitals and University of GenevaGenevaSwitzerland
| | - Silvia Sorce
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Antoine Marteyn
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
- Division of GeriatricsDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
- Division of Internal Medicine for the AgedDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| | - Roberta Ghidoni
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Luisa Benussi
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - François R Herrmann
- Division of GeriatricsDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| | - Karl‐Heinz Krause
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Dina Zekry
- Division of Internal Medicine for the AgedDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| |
Collapse
|
7
|
Pawluk H, Kołodziejska R, Grześk G, Woźniak A, Kozakiewicz M, Kosinska A, Pawluk M, Grześk-Kaczyńska M, Grzechowiak E, Wojtasik J, Kozera G. The Potential Role of RANTES in Post-Stroke Therapy. Cells 2023; 12:2217. [PMID: 37759440 PMCID: PMC10526279 DOI: 10.3390/cells12182217] [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: 08/12/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
One of the key response mechanisms to brain damage, that results in neurological symptoms, is the inflammatory response. It triggers processes that exacerbate neurological damage and create the right environment for the subsequent repair of damaged tissues. RANTES (Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted) chemokine(C-C motif) ligand 5 (CCL5) is one of the chemokines that may have a dual role in stroke progression involving aggravating neuronal damage and playing an important role in angiogenesis and endothelial repair. This study concerned patients with ischemic stroke (AIS), whose CCL5 concentration was measured at various time intervals and was compared with the control group. In addition, the effect of this biomarker on neurological severity and functional prognosis was investigated. Compared to healthy patients, a higher concentration of this chemokine was demonstrated in less than 4.5 h, 24 h and on the seventh day. Differences in CCL5 levels were found to be dependent on the degree of disability and functional status assessed according to neurological scales (modified Rankin Scale, National Institutes of Health Stroke Scale). In addition, differences between various subtypes of stroke were demonstrated, and an increase in CCL5 concentration was proven to be a negative predictor of mortality in patients with AIS. The deleterious effect of CCL5 in the acute phase of stroke and the positive correlation between the tested biomarkers of inflammation were also confirmed.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (A.W.); (M.P.)
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (A.W.); (M.P.)
| | - Grzegorz Grześk
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Ujejskiego 75, 85-168 Bydgoszcz, Poland; (G.G.); (M.G.-K.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (A.W.); (M.P.)
| | - Mariusz Kozakiewicz
- Division of Biochemistry and Biogerontology, Department of Geriatrics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Dębowa 3, 85-626 Bydgoszcz, Poland;
| | - Agnieszka Kosinska
- Centre for Languages & International Education, University College London, 26 Bedford Way, London WC1H 0AP, UK;
| | - Mateusz Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (A.W.); (M.P.)
| | - Magdalena Grześk-Kaczyńska
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Ujejskiego 75, 85-168 Bydgoszcz, Poland; (G.G.); (M.G.-K.)
| | - Elżbieta Grzechowiak
- Department of Neurology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Marii Skłodowskiej Curie 9, 85-094 Bydgoszcz, Poland;
| | - Jakub Wojtasik
- Statistical Analysis Centre, Nicolaus Copernicus University in Toruń, Chopin 12/18, 87-100 Toruń, Poland;
| | - Grzegorz Kozera
- Centre of Medical Simulations, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdańsk, Poland;
| |
Collapse
|
8
|
Jing K, Chen F, Shi X, Guo J, Liu X. Dual effect of C-C motif chemokine receptor 5 on ischemic stroke: More harm than benefit? Eur J Pharmacol 2023:175857. [PMID: 37321471 DOI: 10.1016/j.ejphar.2023.175857] [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: 02/21/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke involves a series of complex pathological mechanisms, of which neuroinflammation is currently the most widely recognized. C-C motif chemokine receptor 5 (CCR5) has recently been shown to be upregulated after cerebral ischemia. Notably, CCR5 is not only involved in neuroinflammation, but also in the blood-brain barrier, neural structures, and connections. Accumulating experimental studies indicate that CCR5 has a dual effect on ischemic stroke. In the acute phase after cerebral ischemia, the pro-inflammatory and disruptive effect of CCR5 on the blood-brain barrier predominates. However, in the chronic phase, the effect of CCR5 on the repair of neural structures and connections is thought to be cell-type dependent. Interestingly, clinical evidence has shown that CCR5 might be harmful rather than beneficial. CCR5-Δ32 mutation or CCR5 antagonist exerts a neuroprotective effect in patients with ischemic stroke. Considering CCR5 as an attractive potential target, we introduce the current research progress of the entangled relationships between CCR5 and ischemic stroke. Clinical data are still needed to determine the efficacy of activating or inactivating CCR5 in the treatment of ischemic stroke, especially for potential phase- or cell type-dependent treatments in the future.
Collapse
Affiliation(s)
- Kai Jing
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Feng Chen
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xiaofei Shi
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jinmin Guo
- Department of Clinical Pharmacy, 960th Hospital of Joint Logistic Support Force, Shandong, Jinan, China.
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China.
| |
Collapse
|
9
|
Cheng Z, Ding Y, Rajah GB, Gao J, Li F, Ma L, Geng X. Vertebrobasilar artery cooling infusion in acute ischemic stroke for posterior circulation following thrombectomy: Rationale, design and protocol for a prospective randomized controlled trial. Front Neurosci 2023; 17:1149767. [PMID: 37113154 PMCID: PMC10126519 DOI: 10.3389/fnins.2023.1149767] [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: 01/23/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Although endovascular mechanical thrombectomy demonstrates clinical efficacy in posterior circulation acute ischemic stroke (AIS), only one third of these patients attain functional independence with a third of patients' expiring despite vascular recanalization. Neuroprotection strategies, such as therapeutic hypothermia (TH) have been considered a promising adjunctive treatment in AIS. We propose the following rationale, design and protocol for a prospective randomized controlled trial (RCT) aimed to determine whether Vertebrobasilar Artery Cooling Infusion (VACI) improves functional outcomes in posterior circulation AIS patients post mechanical thrombectomy. METHODS Subjects in the study will be assigned randomly to either the cooling infusion or the control group in a 1:1 ratio (n = 40). Patients allocated to the cooling infusion group will receive 300 ml cool saline at 4C through the catheter (30 ml/min) into vertebral artery after thrombectomy. The control group will receive the same volume of 37C saline. All patients enrolled will receive standard care according to current guidelines for stroke management. The primary outcome is symptomatic intracranial hemorrhage (ICH), whereas the secondary outcomes include functional outcome score, infarction volume, mortality, ICH, fatal ICH, cerebral vasospasm, coagulation abnormality, pneumonia and urinary infection. DISCUSSIONS This study will determine the preliminary safety, feasibility, and neuroprotective benefits of VACI in posterior circulation AIS patients with reperfusion therapy. The results of this study may provide evidence for VACI as a new therapy in posterior circulation AIS. CLINICAL TRIAL REGISTRATION www.chictr.org.cn, ChiCTR2200065806, registered on November 15, 2022.
Collapse
Affiliation(s)
- Zhe Cheng
- Department of Neurology and Stroke Intervention and Translational Center (SITC), Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Gary B. Rajah
- Department of Neurosurgery, Munson Healthcare, Traverse City, MI, United States
| | - Jie Gao
- Department of Neurology and Stroke Intervention and Translational Center (SITC), Luhe Hospital, Capital Medical University, Beijing, China
| | - Fenghai Li
- Department of Neurology and Stroke Intervention and Translational Center (SITC), Luhe Hospital, Capital Medical University, Beijing, China
| | - Linlin Ma
- Department of Neurology and Stroke Intervention and Translational Center (SITC), Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Department of Neurology and Stroke Intervention and Translational Center (SITC), Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Greco GA, Rock M, Amontree M, Lanfranco MF, Korthas H, Hong SH, Turner RS, Rebeck GW, Conant K. CCR5 deficiency normalizes TIMP levels, working memory, and gamma oscillation power in APOE4 targeted replacement mice. Neurobiol Dis 2023; 179:106057. [PMID: 36878326 PMCID: PMC10291850 DOI: 10.1016/j.nbd.2023.106057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/07/2023] Open
Abstract
The APOE4 allele increases the risk for Alzheimer's disease (AD) in a dose-dependent manner and is also associated with cognitive decline in non-demented elderly controls. In mice with targeted gene replacement (TR) of murine APOE with human APOE3 or APOE4, the latter show reduced neuronal dendritic complexity and impaired learning. APOE4 TR mice also show reduced gamma oscillation power, a neuronal population activity which is important to learning and memory. Published work has shown that brain extracellular matrix (ECM) can reduce neuroplasticity as well as gamma power, while attenuation of ECM can instead enhance this endpoint. In the present study we examine human cerebrospinal fluid (CSF) samples from APOE3 and APOE4 individuals and brain lysates from APOE3 and APOE4 TR mice for levels of ECM effectors that can increase matrix deposition and restrict neuroplasticity. We find that CCL5, a molecule linked to ECM deposition in liver and kidney, is increased in CSF samples from APOE4 individuals. Levels of tissue inhibitor of metalloproteinases (TIMPs), which inhibit the activity of ECM-degrading enzymes, are also increased in APOE4 CSF as well as astrocyte supernatants brain lysates from APOE4 TR mice. Importantly, as compared to APOE4/wild-type heterozygotes, APOE4/CCR5 knockout heterozygotes show reduced TIMP levels and enhanced EEG gamma power. The latter also show improved learning and memory, suggesting that the CCR5/CCL5 axis could represent a therapeutic target for APOE4 individuals.
Collapse
Affiliation(s)
- Griffin A Greco
- Georgetown University School of Medicine (GUMC), Department of Pharmacology, United States of America
| | | | - Matthew Amontree
- GUMC, United States of America; Interdisciplinary Program in Neuroscience, United States of America
| | | | - Holly Korthas
- Interdisciplinary Program in Neuroscience, United States of America
| | - Sung Hyeok Hong
- GUMC, Department of Biochemistry and Molecular & Cellular Biology, United States of America
| | | | - G William Rebeck
- Interdisciplinary Program in Neuroscience, United States of America; GUMC, Department of Neuroscience, United States of America
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, United States of America; GUMC, Department of Neuroscience, United States of America.
| |
Collapse
|
11
|
Lin X, Wang H, Huang S, Chen L, Yang S, Zhao P, Lin Z, Yang J, Ruan L, Ni H, Wang K, Wen M, Jin K, Zhuge Q. A Reliable Nonhuman Primate Model of Ischemic Stroke with Reproducible Infarct Size and Long-term Sensorimotor Deficits. Aging Dis 2023; 14:245-255. [PMID: 36818571 PMCID: PMC9937702 DOI: 10.14336/ad.2022.0722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022] Open
Abstract
A nonhuman primate model of ischemic stroke is considered as an ideal preclinical model to replicate various aspects of human stroke because of their similarity to humans in genetics, neuroanatomy, physiology, and immunology. However, it remains challenging to produce a reliable and reproducible stroke model in nonhuman primates due to high mortality and variable outcomes. Here, we developed a focal cerebral ischemic model induced by topical application of 50% ferric chloride (FeCl3) onto the MCA-M1 segment through a cranial window in the cynomolgus monkeys. We found that FeCl3 rapidly produced a stable intraarterial thrombus that caused complete occlusion of the MCA, leading to the quick decrease of the regional cerebral blood flow in 10 min. A typical cortical infarct was detected 24 hours by magnetic resonance imaging (MRI) and was stable at least for 1 month after surgery. The sensorimotor deficit assessed by nonhuman primate stroke scale was observed at 1 day and up to 3 months after ischemic stroke. No spontaneous revascularization or autolysis of thrombus was observed, and vital signs were not affected. All operated cynomolgus monkeys survived. Our data suggested that FeCl3-induced stroke in nonhuman primates was a replicable and reliable model that is necessary for the correct prediction of the relevance of experimental therapeutic approaches in human beings.
Collapse
Affiliation(s)
- Xiao Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Hua Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Lefu Chen
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Su Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Peiqi Zhao
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Zhongxiao Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Jianjing Yang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Linhui Ruan
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Haoqi Ni
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Kankai Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Min Wen
- Department of Neurology, Guangzhou First People's Hospital, Guangzhou, China.
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.,Correspondence should be addressed to: Dr. Qichuan Zhuge, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China. .
| |
Collapse
|
12
|
CCL5 Levels Predict Stroke Volume Growth in Acute Ischemic Stroke and Significantly Diminish in Hemorrhagic Stroke Patients. Int J Mol Sci 2022; 23:ijms23179967. [PMID: 36077361 PMCID: PMC9456070 DOI: 10.3390/ijms23179967] [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: 07/18/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Stroke remains an important health challenge. Here, we study whether circulating chemokine (C-C motif) ligand 5 (CCL5) levels may predict clinical outcomes for stroke patients. A total of 100 consecutive stroke patients (36 acute ischemic and 64 hemorrhagic) were admitted to the stroke unit. Clinical history data and monitoring parameters were recorded. Blood serum was collected at days 0, 1, and hospital discharge to measure CCL5 levels by ELISA. Infarct or hemorrhagic volume, neurological severity (NIHSS), and functional prognosis (mRankin scale) were measured as clinical outcomes. CCL5 levels were lower in patients with hemorrhagic stroke than in patients with acute ischemic stroke. No differences were found between females and males in both types of stroke. Ischemic stroke patients whose infarct volume grew had lower CCL5 levels at day 0. Levels of CCL5 in ischemic and hemorrhagic patients were not associated with more severe symptoms/worse prognosis (NIHSS > 3; mRankin > 2) at admission or at 3 months. CCL5 could be used as a diagnostic marker to distinguish between ischemic and hemorrhagic strokes. Furthermore, CCL5 levels could predict the infarct volume outcomes in ischemic patients.
Collapse
|
13
|
Feng YQ, Xu ZZ, Wang YT, Xiong Y, Xie W, He YY, Chen L, Liu GY, Li X, Liu J, Wu Q. Targeting C–C Chemokine Receptor 5: Key to Opening the Neurorehabilitation Window After Ischemic Stroke. Front Cell Neurosci 2022; 16:876342. [PMID: 35573839 PMCID: PMC9095921 DOI: 10.3389/fncel.2022.876342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the world’s second major cause of adult death and disability, resulting in the destruction of brain tissue and long-term neurological impairment; induction of neuronal plasticity can promote recovery after stroke. C–C chemokine receptor 5 (CCR5) can direct leukocyte migration and localization and is a co-receptor that can mediate human immunodeficiency virus (HIV) entry into cells. Its role in HIV infection and immune response has been extensively studied. Furthermore, CCR5 is widely expressed in the central nervous system (CNS), is engaged in various physiological activities such as brain development, neuronal differentiation, communication, survival, and learning and memory capabilities, and is also involved in the development of numerous neurological diseases. CCR5 is differentially upregulated in neurons after stroke, and the inhibition of CCR5 in specific regions of the brain promotes motor and cognitive recovery. The mechanism by which CCR5 acts as a therapeutic target to promote neurorehabilitation after stroke has rarely been systematically reported yet. Thus, this review aims to discuss the function of CCR5 in the CNS and the mechanism of its effect on post-stroke recovery by regulating neuroplasticity and the inflammatory response to provide an effective basis for clinical rehabilitation after stroke.
Collapse
|
14
|
Michalettos G, Ruscher K. Crosstalk Between GABAergic Neurotransmission and Inflammatory Cascades in the Post-ischemic Brain: Relevance for Stroke Recovery. Front Cell Neurosci 2022; 16:807911. [PMID: 35401118 PMCID: PMC8983863 DOI: 10.3389/fncel.2022.807911] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/28/2022] [Indexed: 11/28/2022] Open
Abstract
Adaptive plasticity processes are required involving neurons as well as non-neuronal cells to recover lost brain functions after an ischemic stroke. Recent studies show that gamma-Aminobutyric acid (GABA) has profound effects on glial and immune cell functions in addition to its inhibitory actions on neuronal circuits in the post-ischemic brain. Here, we provide an overview of how GABAergic neurotransmission changes during the first weeks after stroke and how GABA affects functions of astroglial and microglial cells as well as peripheral immune cell populations accumulating in the ischemic territory and brain regions remote to the lesion. Moreover, we will summarize recent studies providing data on the immunomodulatory actions of GABA of relevance for stroke recovery. Interestingly, the activation of GABA receptors on immune cells exerts a downregulation of detrimental anti-inflammatory cascades. Conversely, we will discuss studies addressing how specific inflammatory cascades affect GABAergic neurotransmission on the level of GABA receptor composition, GABA synthesis, and release. In particular, the chemokines CXCR4 and CX3CR1 pathways have been demonstrated to modulate receptor composition and synthesis. Together, the actual view on the interactions between GABAergic neurotransmission and inflammatory cascades points towards a specific crosstalk in the post-ischemic brain. Similar to what has been shown in experimental models, specific therapeutic modulation of GABAergic neurotransmission and inflammatory pathways may synergistically promote neuronal plasticity to enhance stroke recovery.
Collapse
Affiliation(s)
- Georgios Michalettos
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- LUBIN Lab—Lunds Laboratorium för Neurokirurgisk Hjärnskadeforskning, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- *Correspondence: Karsten Ruscher
| |
Collapse
|
15
|
Deng S, Gan L, Liu C, Xu T, Zhou S, Guo Y, Zhang Z, Yang GY, Tian H, Tang Y. Roles of Ependymal Cells in the Physiology and Pathology of the Central Nervous System. Aging Dis 2022; 14:468-483. [PMID: 37008045 PMCID: PMC10017161 DOI: 10.14336/ad.2022.0826-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
Ependymal cells are indispensable components of the central nervous system (CNS). They originate from neuroepithelial cells of the neural plate and show heterogeneity, with at least three types that are localized in different locations of the CNS. As glial cells in the CNS, accumulating evidence demonstrates that ependymal cells play key roles in mammalian CNS development and normal physiological processes by controlling the production and flow of cerebrospinal fluid (CSF), brain metabolism, and waste clearance. Ependymal cells have been attached to great importance by neuroscientists because of their potential to participate in CNS disease progression. Recent studies have demonstrated that ependymal cells participate in the development and progression of various neurological diseases, such as spinal cord injury and hydrocephalus, raising the possibility that they may serve as a potential therapeutic target for the disease. This review focuses on the function of ependymal cells in the developmental CNS as well as in the CNS after injury and discusses the underlying mechanisms of controlling the functions of ependymal cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yaohui Tang
- Correspondence should be addressed to: Dr. Yaohui Tang, Med-X Research Institute and School of Biomedical Engineering Shanghai Jiao Tong University, Shanghai, China. .
| |
Collapse
|
16
|
Korzhevskii DE, Tsyba DL, Kirik OV, Alekseeva OS. A Comparison of Microglia Detection in Mammals and Humans Using Purinergic Receptor P2Y12 Labeling. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s002209302105001x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|