1
|
Qi H, Zhang X, Zhang Z, Gao Y, Tian D, Zhao G, Xie Z, Zeng J, Zhang L, Zeng N, Yang R. The extract of chrysanthemum flos mitigates post-stroke sarcopenia by inhibiting PANoptosis and restoring muscle homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156784. [PMID: 40311590 DOI: 10.1016/j.phymed.2025.156784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Sarcopenia and muscle weakness are prevalent complications of ischemic stroke (IS), with limited pharmacological options. This study identifies high-dose extracts of Chrysanthemum Flos (ECF) as a potential therapy for post-stroke muscle dysfunction by targeting PANoptosis-a pro-inflammatory programmed cell death pathway. Through its anti-inflammatory and antioxidant properties, ECF attenuates muscle atrophy and enhances functional recovery, offering novel insights into ISS treatment. PURPOSE To evaluate the therapeutic efficacy of high-dose ECF in ischemic stroke-induced sarcopenia (ISS) and elucidate its regulatory role in PANoptosis-mediated muscle degeneration and protein homeostasis. METHODS Preparation of a rat middle cerebral artery occlusion (MCAO) model using intravascular wire thrombus blockade. Cerebral injury was assessed using laser speckle contrast imaging, triphenyltetrazolium chloride (TTC) staining, and Zea-Longa neurological scoring. ECF's effects on muscle function were evaluated through gait analysis, muscle morphology (length and weight), grip strength, electromyography, and H&E staining. RNA sequencing was conducted to elucidate transcriptomic alterations and enriched pathways associated with ECF in ISS. PANoptosis-mediated myofiber and L6 cell damage was analyzed by flow cytometry (FC), immunofluorescence (IF), immunohistochemistry (IHC), and western blotting (WB). ECF composition and quality were validated using liquid chromatography-mass spectrometry (LC-MS). RESULTS ISS rats showed 83 % reductions in endurance, grip strength, and EMG signals compared to sham (p < 0.01), which improved to 70 % of normal after ECF treatment. ECF significantly increased muscle fiber area, alleviated mitochondrial damage, and improved sarcomere structure (p < 0.001). RNA-seq identified TNF signaling and PANoptosis (apoptosis, pyroptosis, necroptosis) as key drivers of ISS-induced muscle injury. The TNF-targeted inhibitor R7050 further confirmed TNF-α as a critical activator of Z-DNA binding protein 1 (ZBP1). ECF treatment significantly reduced tissue inflammation (p < 0.01) and inhibited ZBP1 expression (p < 0.01). Following ISS, key PANoptosis-related proteins, including ZBP1, Gasdermin D N-terminal fragment (GSDMD-N), Cleaved-Caspase3, Caspase6, Caspase8, phosphorylated mixed-lineage kinase domain-like (p-MLKL), Phosphorylated Receptor-Interacting Protein Kinase 1 (p-RIPK1), Phosphorylated Receptor-Interacting Protein Kinase 3 (p-RIPK3), and NOD-like receptor family pyrin domain containing 3 (NLRP3), were significantly upregulated (p < 0.05), while ECF-H treatment significantly suppressed their expression (p < 0.05, p < 0.01). Additionally, ECF significantly promoted the expression of muscle protein synthesis factors (myogenic differentiation 1 (MyoD1) and recombinant myosin heavy chain 1 (MYH), p < 0.01) and inhibited protein degradation factors (muscle RING-finger protein-1 (MuRF1) and muscle atrophy F-Box protein (MAFbx), p < 0.01), thus maintaining muscle protein homeostasis. The results from PCR, WB, IHC, IF, and FC experiments were consistent with RNA-seq findings. CONCLUSIONS ECF ameliorates ISS in MCAO rats by inhibiting muscle PANoptosis, which simultaneously reduces protein degradation and enhances protein synthesis.
Collapse
Affiliation(s)
- Hu Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Xiongwei Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zeyang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yuanlin Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Tian
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Ge Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zhiqiang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihong Zhang
- Department of Otorhinolaryngology, Chengdu Xinjin District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| | - Ruocong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| |
Collapse
|
2
|
Roberts L, Coutts G, Dickie BR, Smith CJ, South K, Allan SM. Comparison of the Novel Thrombolytic Constitutively Active ADAMTS13 With Clinical Thrombolytics in a Murine Stroke Model. Stroke 2025; 56:1589-1595. [PMID: 40171654 PMCID: PMC12101888 DOI: 10.1161/strokeaha.125.050848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND r-tPA (recombinant tissue-type plasminogen activator) and its variant, TNK (tenecteplase), are the currently approved thrombolytic drugs for the treatment of acute ischemic stroke, but they are ineffective in a proportion of patients due to r-tPA resistance of platelet-rich thrombi. A novel thrombolytic, caADAMTS13 (constitutively active a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) has been shown to improve experimental stroke outcomes where platelet-rich thrombi are present but have not been directly compared with r-tPA or TNK. METHODS We conducted a direct comparison of caADAMTS13 versus r-tPA versus TNK versus vehicle control in the ferric chloride-mediated distal middle cerebral artery occlusion model in mice, which features platelet and VWF (von Willebrand Factor)-rich thrombi that reproduce r-tPA-resistant occlusion. Treatments were administered intravenously 1 hour after ferric chloride application by bolus injection or bolus followed by infusion, as translationally applicable. Laser speckle contrast imaging measured early reperfusion over the hour following treatment, and magnetic resonance imaging measured cerebral blood flow and lesion volume at 24 hours. RESULTS Reperfusion 1 hour after treatment was greatest in caADAMTS13-treated animals. Later cerebral blood flow, 24 hours post-treatment, within the stroke-affected hypoperfused area was higher in caADAMTS13 and r-tPA but not TNK-treated mice. Functionally, this led to the absence of an initial behavioral deficit in caADAMTS13-treated mice, alongside a smaller lesion volume at 24 hours and reduced extent of bleeding. CONCLUSIONS These findings demonstrate an overall suggestion that caADAMTS13 has improved thrombolytic efficacy, compared with current stroke treatments, against platelet-rich thrombi, for which there is currently an unmet clinical need.
Collapse
Affiliation(s)
- Lucy Roberts
- Division of Neuroscience, School of Biological Sciences (L.R., G.C., K.S., S.M.A.), Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, United Kingdom (L.R., G.C., B.R.D., C.J.S., K.S., S.M.A.)
| | - Graham Coutts
- Division of Neuroscience, School of Biological Sciences (L.R., G.C., K.S., S.M.A.), Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, United Kingdom (L.R., G.C., B.R.D., C.J.S., K.S., S.M.A.)
| | - Ben R. Dickie
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences (B.R.D.), Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, United Kingdom (L.R., G.C., B.R.D., C.J.S., K.S., S.M.A.)
| | - Craig J. Smith
- Division of Cardiovascular Sciences, School of Medical Sciences (C.J.S.), Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, United Kingdom (L.R., G.C., B.R.D., C.J.S., K.S., S.M.A.)
- Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, United Kingdom (C.J.S.)
| | - Kieron South
- Division of Neuroscience, School of Biological Sciences (L.R., G.C., K.S., S.M.A.), Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, United Kingdom (L.R., G.C., B.R.D., C.J.S., K.S., S.M.A.)
| | - Stuart M. Allan
- Division of Neuroscience, School of Biological Sciences (L.R., G.C., K.S., S.M.A.), Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, United Kingdom (L.R., G.C., B.R.D., C.J.S., K.S., S.M.A.)
| |
Collapse
|
3
|
Ren Z, Shao F, Chen S, Sun Y, Ding Z, Dong L, Zhang J, Zang Y. Contribution of alterations in peritubular capillary density and microcirculation to the progression of tubular injury and kidney fibrosis. J Pathol 2025; 266:95-108. [PMID: 40103536 DOI: 10.1002/path.6414] [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: 07/15/2024] [Revised: 12/18/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
Peritubular capillary (PTC) rarefaction is a common pathological feature of chronic kidney disease (CKD). The critical function of PTCs in maintaining blood supply for tubular epithelial cells renders PTCs a promising therapeutic target. However, the role of PTC rarefaction in the progression of kidney fibrosis remains elusive. In this study, we first characterized mice with altered PTC density. CD31 staining, together with microvascular network perfusion with FITC-labelled albumin and laser speckle contrast imaging, revealed a significant increase in PTC density in Flt1 heterozygous-deficient mice, whereas homozygous disruption of the plasminogen activator, urokinase receptor gene (Plaur/uPAR), led to a notable decrease in PTC density. Using these genetically distinct mice, we showed that preexisting higher PTC density protected against tubular injury and attenuated the progression of tubulointerstitial fibrosis in two distinct kidney injury models, namely, ischemia-reperfusion injury (IRI) and unilateral ureteral obstruction (UUO). By contrast, Plaur-deficient mice with established lower PTC density displayed exacerbated tubular injury and renal fibrosis when subjected to IRI or UUO. The pathophysiological significance of PTC density was associated with protective effects on tubular cell apoptosis and concomitant regeneration. Finally, vasodilation of the renal capillary with minoxidil, a clinically available drug, effectively prevented UUO-induced tubular injury and renal fibrosis. Moreover, minoxidil treatment abolished the detrimental effect of Plaur deficiency on the UUO-treated kidney, thus suggesting a causative role of PTC density in the susceptibility of Plaur knockout mice to tubular injury following fibrosis. Our results provide an overview of the pathologic significance of PTC density alterations in the progression of CKD, and show that improving peritubular microcirculation is effective in preventing tubular injury and the subsequent renal fibrosis. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Fang Shao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Shuli Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Yanyan Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| |
Collapse
|
4
|
Luka N, South K, Jones R, Unsworth AJ, Coutts G, Mosneag I, Younas M, Bradley A, Wong SY, Collins E, Quigley C, Knight SB, McColl BW, McCulloch L, Grainger JR, Smith CJ, Allan SM. The Role of the VWF/ADAMTS13 Axis in the Thromboinflammatory Response in Ischemic Stroke After SARS-CoV2 Infection. Brain Behav 2025; 15:e70348. [PMID: 39972966 PMCID: PMC11839761 DOI: 10.1002/brb3.70348] [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: 03/20/2024] [Revised: 10/22/2024] [Accepted: 02/02/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND SARS-CoV2 infections increase the risk of ischemic stroke (IS), potentially through a thromboinflammatory cascade driven by an imbalance in the ratio of Von Willebrand Factor (VWF) and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), leading to the formation of ultra-large VWF (UL-VWF). However, the SARS-CoV2 infection's contribution to any VWF/ADAMTS13 axis imbalance and the subsequent thromboinflammatory response post-stroke remain poorly understood. METHODS We performed a detailed thromboinflammatory profile of the plasma samples from three experimental cohorts matched by age, sex, and stroke severity: non-stroke controls (n = 23), SARS-CoV2 negative IS (n = 22), and SARS-CoV2 positive IS (n = 24). SARS-CoV2 positive IS patients presented varying degrees of infection severity. RESULTS We observed an increase in VWF and UL-VWF and a decrease in ADAMTS13 in the SARS-CoV2 positive IS cohort, suggesting a VWF/ADAMTS13 axis imbalance. Interleukin-6 (IL-6) levels were positively correlated with VWF and negatively correlated with ADAMTS13, suggesting that IL-6 may drive this imbalance. Fibrinogen and D-Dimers were elevated in SARS-CoV2 negative IS cohort and SARS-CoV2 positive IS cohort, but D-Dimers were within the normal range, indicating no disseminated intravascular coagulation. Factor IX (FIX) was elevated in the SARS-CoV2 negative IS cohort. Tissue plasminogen activator (tPA) was elevated in the SARS-CoV2 positive IS cohort, suggesting no fibrinolysis defects. Matrix Metalloproteinase-2 (MMP-2) and soluble Intracellular Adhesion Molecule-1 (sICAM-1) were elevated in the SARS-CoV2 negative IS cohort. CONCLUSIONS We show that SARS-CoV2 infections drive a VWF/ADAMTS13 axis imbalance, inducing an increase in tPA while decreasing FIX, MMP-2, and sICAM-1 post-stroke.
Collapse
Affiliation(s)
- Nadim Luka
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Kieron South
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Rachel Jones
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of Cardiovascular SciencesThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
| | - Amanda J. Unsworth
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Graham Coutts
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Ioana‐Emilia Mosneag
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
| | - Mehwish Younas
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
| | - Amy Bradley
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Siew Yan Wong
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Ellen Collins
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Chloe Quigley
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Sean B. Knight
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
- Division of Immunology, Immunity to Infection and Respiratory MedicineThe University of ManchesterManchesterUK
| | - Barry W. McColl
- UK Dementia Research Institute, Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - Laura McCulloch
- Centre for Inflammation Research, Institute for Regeneration and RepairThe University of EdinburghEdinburghUK
| | - John R. Grainger
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
- Division of Immunology, Immunity to Infection and Respiratory MedicineThe University of ManchesterManchesterUK
| | - Craig J. Smith
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of Cardiovascular SciencesThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of NeuroscienceThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthManchester Academic Health Science Centre, The University of ManchesterManchesterUK
| |
Collapse
|
5
|
Luo Y, Dong W, Yuan L, Zhu YA, Zhang DD, Ni H, Zhu W. The Role of Thrombo-inflammation in Ischemic Stroke: Focus on the Manipulation and Clinical Application. Mol Neurobiol 2025; 62:2362-2375. [PMID: 39107669 DOI: 10.1007/s12035-024-04397-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/22/2024] [Indexed: 01/28/2025]
Abstract
Stroke leaves a great economic burden due to its high morbidity and mortality. Rapid revascularization of targeted vessel(s) is the effective treatment for ischemic stroke, but subsequent ischemia-reperfusion (I/R) injury is a common complication following revascularization, leading to microcirculation dysfunction and infarct volume increase. Thrombo-inflammation, the interaction between thrombosis and inflammation, plays a critical role in the pathophysiology of ischemic stroke. In the context of I/R injury, thrombo-inflammation consists of platelet activation, endothelial injury, and inflammatory cell infiltration. Numerous studies are devoted to exploring methods of regulating thrombo-inflammation to mitigate I/R injury post-stroke, including blocking activations of platelets and neutrophils. Drugs such as antiplatelet medications, anticoagulants, and glucocorticoids have been confirmed to have the potential to regulate thrombo-inflammation. Furthermore, several recently developed drugs have also shown promises in relieving I/R injury by manipulating thrombo-inflammation. However, the majority of these studies are still in the preclinical stage. Herein, in this review, we will address the mechanisms of thrombo-inflammation in ischemic stroke, related research advances, and particularly the clinical feasibility of thrombo-inflammation as a therapeutic strategy against I/R injury.
Collapse
Affiliation(s)
- Yuanfei Luo
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weichen Dong
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Linying Yuan
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunqing Amelia Zhu
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
| | - Dachuan Dustin Zhang
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, M5G 2M1, Canada
- CCOA Therapeutics Inc., Toronto, ON, M5B 1W8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Wusheng Zhu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
6
|
Evans LE, Gray AL, Walsh KR, Danby TGE, Pritchard HAT, Allan SM, Gurney AM, Greenstein AS, Schiessl I. Combining In Vivo Two-Photon and Laser Speckle Microscopy With the Ex Vivo Capillary-Parenchymal Arteriole Preparation as a Novel Approach to Study Neurovascular Coupling. Microcirculation 2025; 32:e70001. [PMID: 39777814 PMCID: PMC11706670 DOI: 10.1111/micc.70001] [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: 02/14/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE Cerebral blood flow (CBF) decline is increasingly recognized as an area of importance for targeting neurodegenerative disorders, yet full understanding of the mechanisms that underlie CBF changes are lacking. Animal models are crucial for expanding our knowledge as methods for studying global CBF and neurovascular coupling in humans are limited and require expensive specialized scanners. METHODS Use of appropriate animal models can increase our understanding of cerebrovascular function, so we have combined chronic cranial windows with in vivo two-photon and laser speckle microscopy and ex vivo capillary-parenchymal arteriole (CaPA) preparations. Chronic cranial windows allow for longitudinal direct observation of the cerebral microvasculature and surrounding parenchyma while the CaPA preparation can assess capillary and arteriole function in isolation of the neuronal tissue. RESULTS Here, we found that extra-dural cranial windows and related imaging protocols do not affect vascular function in the CaPA preparation. Cortical vessels from animals that have undergone imaging can therefore be taken to discover physiological alterations in the cerebral vasculature that contribute to any observed in vivo changes. CONCLUSION This approach will enhance neurodegenerative research with the benefit of limiting animal usage.
Collapse
Affiliation(s)
- Lowri E. Evans
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Anna L. Gray
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Wellcome Centre for Cell‐Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Katy R. Walsh
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Thea G. E. Danby
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Harry A. T. Pritchard
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Alison M. Gurney
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Adam S. Greenstein
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
7
|
Sun A, Huang W, Jin K, Zhong M, Yu B, Li X, Wang Y, Liu H. A multiple targeting rapamycin and SS31 conjugate enhances ischemic stroke therapy. Expert Opin Drug Deliv 2025; 22:109-120. [PMID: 39663652 DOI: 10.1080/17425247.2024.2440094] [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: 05/26/2024] [Accepted: 11/16/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND The identification of drugs targeting multiple pathways is essential for comprehensive protection against cerebral ischemia-reperfusion injury. RESEARCH DESIGN AND METHODS This study aimed to develop RS31, a multi-target cytoprotectant composed of SS31 (an oxidative stress mitigator) and rapamycin (Rapa), contributes anti-inflammatory and blood-brain barrier protection. RS31 was synthesized using click chemistry, and its ability to scavenge reactive oxygen species (ROS) and reduce inflammation was tested in H2O2-injured PC12 cells and LPS-stimulated BV2 cells. A C57BL/6 mouse model of transient middle cerebral artery occlusion/reperfusion (tMCAO/R) was established to assess the effect of RS31 on inflammatory factors in ischemic brain tissue. Finally, the potential of combining RS31 with PLGA microparticles (MPs) to further reduce brain edema was investigated. RESULTS RS31 effectively scavenged ROS and reduced inflammation. It showed a ~ 4-fold higher concentration in cerebral ischemic regions, significant reducing infarction and improving neurological function. RS31 also effectively reduced inflammatory factors, lowered malondialdehyde (MDA) levels, and increased superoxide dismutase (SOD) activity, showing strong efficacy in treating ischemic stroke. CONCLUSIONS In vivo delivery of RS31 is an effective therapeutic strategy for I/R injury, providing a general framework for developing multi-targeted drugs against inflammatory diseases and excessive ROS production.
Collapse
Affiliation(s)
- Andi Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Weijia Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Kai Jin
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Mingyuan Zhong
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Bohong Yu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
8
|
South K, Roberts L, Gray A, Luka N, Strangward P, Coutts G, Smith CJ, Schiessl I, Allan SM. Inhibition of neutrophil rolling and migration by caADAMTS13 in vitro and in mouse models of thrombosis and inflammation. Biomed Pharmacother 2024; 178:117166. [PMID: 39029401 DOI: 10.1016/j.biopha.2024.117166] [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: 05/02/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024] Open
Abstract
Recent investigation of a constitutively active ADAMTS13 variant (caADAMTS13) in murine models of acute ischaemic stroke (AIS) have revealed a potential anti-inflammatory mechanism of action contributing to its protective effect. However, it remains unclear whether these observations are a direct result of VWF proteolysis by caADAMTS13. We have implemented state of the art in vitro assays of neutrophil rolling and transmigration to quantify the impact of caADAMTS13 on these processes. Moreover, we have tested caADAMTS13 in two in vivo assays of neutrophil migration to confirm the impact of the treatment on the neutrophil response to sterile inflammation. Neutrophil rolling, over an interleukin-1β stimulated hCMEC/D3 monolayer, is directly inhibited by caADAMTS13, reducing the proportion of neutrophils rolling to 9.5 ± 3.8 % compared to 18.0 ± 4.5 % in untreated controls. Similarly, neutrophil transmigration recorded in real-time, was significantly suppressed in the presence of caADAMTS13 which reduced the number of migration events to a level like that in unstimulated controls (18.0 ± 4.5 and 15.8 ± 7.5 cells/mm2/h, respectively). Brain tissue from mice undergoing experimental focal cerebral ischaemia has indicated the inhibition of this process by caADAMTS13. This is supported by caADAMTS13's ability to reduce neutrophil migration into the peritoneal cavity in an ischaemia-independent model of sterile inflammation, with the VWF-dependent mechanism by which this occurs being confirmed using a second experimental stroke model. These findings will be an important consideration in the further development of caADAMTS13 as a potential therapy for AIS and other thromboinflammatory pathologies, including cardiovascular disease.
Collapse
Affiliation(s)
- Kieron South
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK.
| | - Lucy Roberts
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Anna Gray
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Nadim Luka
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Patrick Strangward
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Graham Coutts
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Craig J Smith
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK; Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - Ingo Schiessl
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Liu X, Wang J, Jin J, Hu Q, Zhao T, Wang J, Gao J, Man J. S100A9 deletion in microglia/macrophages ameliorates brain injury through the STAT6/PPARγ pathway in ischemic stroke. CNS Neurosci Ther 2024; 30:e14881. [PMID: 39107960 PMCID: PMC11303267 DOI: 10.1111/cns.14881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/30/2024] [Accepted: 07/14/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Microglia and infiltrated macrophages (M/M) are integral components of the innate immune system that play a critical role in facilitating brain repair after ischemic stroke (IS) by clearing cell debris. Novel therapeutic strategies for IS therapy involve modulating M/M phenotype shifting. This study aims to elucidate the pivotal role of S100A9 in M/M and its downstream STAT6/PPARγ signaling pathway in neuroinflammation and phagocytosis after IS. METHODS In the clinical study, we initially detected the expression pattern of S100A9 in monocytes from patients with acute IS and investigated its association with the long-term prognosis. In the in vivo study, we generated the S100A9 conditional knockout (CKO) mice and compared the stroke outcomes with the control group. We further tested the S100A9-specific inhibitor paqunimod (PQD), for its pharmaceutical effects on stroke outcomes. Transcriptomics and in vitro studies were adopted to explore the mechanism of S100A9 in modulating the M/M phenotype, which involves the regulation of the STAT6/PPARγ signaling pathway. RESULTS S100A9 was predominantly expressed in classical monocytes and was correlated with unfavorable outcomes in patients of IS. S100A9 CKO mitigated infarction volume and white matter injury, enhanced cerebral blood flow and functional recovery, and prompted anti-inflammation phenotype and efferocytosis after tMCAO. The STAT6/PPARγ pathway, an essential signaling cascade involved in immune response and inflammation, might be the downstream target mediated by S100A9 deletion, as evidenced by the STAT6 phosphorylation inhibitor AS1517499 abolishing the beneficial effect of S100A9 inhibition in tMCAO mice and cell lines. Moreover, S100A9 inhibition by PQD treatment protected against neuronal death in vitro and brain injuries in vivo. CONCLUSION This study provides evidence for the first time that S100A9 in classical monocytes could potentially be a biomarker for predicting IS prognosis and reveals a novel therapeutic strategy for IS. By demonstrating that S100A9-mediated M/M polarization and phagocytosis can be reversed by S100A9 inhibition in a STAT6/PPARγ pathway-dependent manner, this study opens up new avenues for drug development in the field.
Collapse
Affiliation(s)
- Xi Liu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Jian Jin
- MRI imaging core, Medical Research CenterThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Qiongqiong Hu
- Department of Neurology, Zhengzhou Central HospitalZhengzhou UniversityZhengzhouChina
| | - Ting Zhao
- Department of NeurologyPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Jianbo Gao
- Department of RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiang Man
- Department of RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
10
|
Ceulemans A, Spronk HMH, Ten Cate H, van Zwam WH, van Oostenbrugge RJ, Nagy M. Current and potentially novel antithrombotic treatment in acute ischemic stroke. Thromb Res 2024; 236:74-84. [PMID: 38402645 DOI: 10.1016/j.thromres.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/22/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Acute ischemic stroke (AIS) is the most common type of stroke and requires immediate reperfusion. Current acute reperfusion therapies comprise the administration of intravenous thrombolysis and/or endovascular thrombectomy. Although these acute reperfusion therapies are increasingly successful, optimized secondary antithrombotic treatment remains warranted, specifically to reduce the risk of major bleeding complications. In the development of AIS, coagulation and platelet activation play crucial roles by driving occlusive clot formation. Recent studies implicated that the intrinsic route of coagulation plays a more prominent role in this development, however, this is not fully understood yet. Next to the acute treatments, antithrombotic therapy, consisting of anticoagulants and/or antiplatelet therapy, is successfully used for primary and secondary prevention of AIS but at the cost of increased bleeding complications. Therefore, better understanding the interplay between the different pathways involved in the pathophysiology of AIS might provide new insights that could lead to novel treatment strategies. This narrative review focuses on the processes of platelet activation and coagulation in AIS, and the most common antithrombotic agents in primary and secondary prevention of AIS. Furthermore, we provide an overview of promising novel antithrombotic agents that could be used to improve in both acute treatment and stroke prevention.
Collapse
Affiliation(s)
- Angelique Ceulemans
- Department of Neurology, Maastricht University Medical Center+, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Henri M H Spronk
- School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Biochemistry, Maastricht University Medical Center+, Maastricht, the Netherlands; Thrombosis Expertise Center, Heart & Vascular Center, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Hugo Ten Cate
- School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands; Department of internal medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Thrombosis Expertise Center, Heart & Vascular Center, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Wim H van Zwam
- School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- Department of Neurology, Maastricht University Medical Center+, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Magdolna Nagy
- School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Biochemistry, Maastricht University Medical Center+, Maastricht, the Netherlands; Thrombosis Expertise Center, Heart & Vascular Center, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
11
|
Sri S, Greenstein A, Granata A, Collcutt A, Jochems ACC, McColl BW, Castro BD, Webber C, Reyes CA, Hall C, Lawrence CB, Hawkes C, Pegasiou-Davies CM, Gibson C, Crawford CL, Smith C, Vivien D, McLean FH, Wiseman F, Brezzo G, Lalli G, Pritchard HAT, Markus HS, Bravo-Ferrer I, Taylor J, Leiper J, Berwick J, Gan J, Gallacher J, Moss J, Goense J, McMullan L, Work L, Evans L, Stringer MS, Ashford MLJ, Abulfadl M, Conlon N, Malhotra P, Bath P, Canter R, Brown R, Ince S, Anderle S, Young S, Quick S, Szymkowiak S, Hill S, Allan S, Wang T, Quinn T, Procter T, Farr TD, Zhao X, Yang Z, Hainsworth AH, Wardlaw JM. A multi-disciplinary commentary on preclinical research to investigate vascular contributions to dementia. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100189. [PMID: 37941765 PMCID: PMC10628644 DOI: 10.1016/j.cccb.2023.100189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 11/10/2023]
Abstract
Although dementia research has been dominated by Alzheimer's disease (AD), most dementia in older people is now recognised to be due to mixed pathologies, usually combining vascular and AD brain pathology. Vascular cognitive impairment (VCI), which encompasses vascular dementia (VaD) is the second most common type of dementia. Models of VCI have been delayed by limited understanding of the underlying aetiology and pathogenesis. This review by a multidisciplinary, diverse (in terms of sex, geography and career stage), cross-institute team provides a perspective on limitations to current VCI models and recommendations for improving translation and reproducibility. We discuss reproducibility, clinical features of VCI and corresponding assessments in models, human pathology, bioinformatics approaches, and data sharing. We offer recommendations for future research, particularly focusing on small vessel disease as a main underpinning disorder.
Collapse
Affiliation(s)
- Sarmi Sri
- UK Dementia Research Institute Headquarters, 6th Floor Maple House, London W1T 7NF, UK
| | - Adam Greenstein
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Alessandra Granata
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Alex Collcutt
- UK Dementia Research Institute Headquarters, 6th Floor Maple House, London W1T 7NF, UK
| | - Angela C C Jochems
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Barry W McColl
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Blanca Díaz Castro
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Caleb Webber
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, UK
| | - Carmen Arteaga Reyes
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Catherine Hall
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Catherine B Lawrence
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cheryl Hawkes
- Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | | | - Claire Gibson
- School of Psychology, University of Nottingham, Nottingham NG7 2UH, UK
| | - Colin L Crawford
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, UNICAEN, INSERM UMR-S U1237, , GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Department of clinical research, Caen-Normandie University Hospital, Caen, France
| | - Fiona H McLean
- Division of Systems Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Frances Wiseman
- UK Dementia Research Institute, University College London, London WC1N 3BG, UK
| | - Gaia Brezzo
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Giovanna Lalli
- UK Dementia Research Institute Headquarters, 6th Floor Maple House, London W1T 7NF, UK
| | - Harry A T Pritchard
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Hugh S Markus
- Stroke Research Group, Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Isabel Bravo-Ferrer
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Jade Taylor
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James Leiper
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jason Berwick
- Department of Psychology, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Jian Gan
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - John Gallacher
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - Jonathan Moss
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, UK
| | - Jozien Goense
- Neuroscience Program, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Department of Psychology, University of Illinois, Urbana-Champaign, Champaign, IL, USA
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- School of Psychology and Neuroscience, University of Glasgow, UK
| | - Letitia McMullan
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Lorraine Work
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow; Glasgow; UK
| | - Lowri Evans
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Michael S Stringer
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - MLJ Ashford
- Division of Systems Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Mohamed Abulfadl
- Dementia Research Group, Department of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol BS10 5NB, UK
| | - Nina Conlon
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Paresh Malhotra
- Department of Brain Sciences, Imperial College London, London, UK
- Department of Neurology, Imperial College Healthcare NHS Trust, London, UK
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London and the University of Surrey, UK
| | - Philip Bath
- Stroke Trials Unit, University of Nottingham, Nottingham, UK; Stroke, Medicine Division, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Rebecca Canter
- Dementia Discovery Fund, SV Health Managers LLP, London, UK
| | - Rosalind Brown
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Selvi Ince
- Dementia Research Group, Department of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol BS10 5NB, UK
| | - Silvia Anderle
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
- Department of Neuroscience, Physiology and Pharmacology, University College London, UK
| | - Simon Young
- Dementias Platform UK, Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Sophie Quick
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Stefan Szymkowiak
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, UK
| | - Steve Hill
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, UK
| | - Stuart Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Tao Wang
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Evolution, Infection and Genomic Sciences, Faculty of Biology Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Terry Quinn
- College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - Tessa Procter
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, UK
| | - Tracy D Farr
- School of Life Sciences, Physiology, Pharmacology, and Neuroscience Division, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Xiangjun Zhao
- Division of Evolution, Infection and Genomic Sciences, Faculty of Biology Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Zhiyuan Yang
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Atticus H Hainsworth
- Molecular and Clinical Sciences Research Institute, St George's University of London SW17 0RE, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Sakai K, Matsumoto M. Clinical Manifestations, Current and Future Therapy, and Long-Term Outcomes in Congenital Thrombotic Thrombocytopenic Purpura. J Clin Med 2023; 12:3365. [PMID: 37240470 PMCID: PMC10219024 DOI: 10.3390/jcm12103365] [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: 02/28/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Congenital thrombotic thrombocytopenic purpura (cTTP) is an extremely rare disease characterized by the severe deficiency of a disintegrin and metalloproteinase with thrombospondin type 1 motifs 13 (ADAMTS13), caused by ADAMTS13 mutations. While ADAMTS13 supplementation by fresh frozen plasma (FFP) infusion immediately corrects platelet consumption and resolves thrombotic symptoms in acute episodes, FFP treatment can lead to intolerant allergic reactions and frequent hospital visits. Up to 70% of patients depend on regular FFP infusions to normalize their platelet counts and avoid systemic symptoms, including headache, fatigue, and weakness. The remaining patients do not receive regular FFP infusions, mainly because their platelet counts are maintained within the normal range or because they are symptom-free without FFP infusions. However, the target peak and trough levels of ADAMTS13 to prevent long-term comorbidity with prophylactic FFP and the necessity of treating FFP-independent patients in terms of long-term clinical outcomes are yet to be determined. Our recent study suggests that the current volumes of FFP infusions are insufficient to prevent frequent thrombotic events and long-term ischemic organ damage. This review focuses on the current management of cTTP and its associated issues, followed by the importance of upcoming recombinant ADAMTS13 therapy.
Collapse
Affiliation(s)
- Kazuya Sakai
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara 634-8522, Japan;
| | - Masanori Matsumoto
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara 634-8522, Japan;
- Department of Hematology, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
13
|
VWF-targeted thrombolysis to overcome rh-tPA resistance in experimental murine ischemic stroke models. Blood 2022; 140:2844-2848. [PMID: 35960811 DOI: 10.1182/blood.2022016342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/01/2022] [Accepted: 07/26/2022] [Indexed: 01/05/2023] Open
Abstract
Recombinant human tissue plasminogen activator (rh-tPA) is an important thrombolytic agent for treatment of acute ischemic stroke. It requires fibrin binding for plasminogen activation. In contrast, Microlyse, a novel thrombolytic agent, requires von Willebrand factor (VWF) binding for plasminogen activation. We compared rh-tPA with Microlyse, administered 20 minutes after inducing thrombosis, in 2 randomized blinded acute ischemic stroke mouse models. Thrombosis was induced in the middle cerebral artery with different experimental triggers. Where thrombin infusion generates fibrin-rich thrombi, topical FeCl3 application generates platelet-rich thrombi. In the fibrin-rich model, both rh-tPA and Microlyse increased cortical reperfusion (determined by laser speckle imaging) 10 minutes after therapy administration (35.8 ± 17.1%; P = .001 39.3 ± 13.1%; P < .0001; 15.6 ± 7.5%, respectively, vs vehicle). In addition, both thrombolytic agents reduced cerebral lesion volume (determined by magnetic resonance imaging) after 24 hours (18.9 ± 11.2 mm3; P = .033; 16.1 ± 13.9 mm3; P = .018; 26.6 ± 5.6 mm3, respectively, vs vehicle). In the platelet-rich model, neither rh-tPA nor Microlyse increased cortical reperfusion 10 minutes after therapy (7.6 ± 8.8%; P = .216; 16.3 ± 13.9%; P = .151; 10.1 ± 7.9%, respectively, vs vehicle). However, Microlyse, but not rh-tPA, decreased cerebral lesion volumes (13.9 ± 11.4 mm3; P < .001; 23.6 ± 11.1 mm3; P = .188; 30.3 ± 10.9 mm3, respectively, vs vehicle). These findings support broad applicability of Microlyse in ischemic stroke, irrespective of the thrombus composition.
Collapse
|
14
|
DeYoung V, Singh K, Kretz CA. Mechanisms of ADAMTS13 regulation. J Thromb Haemost 2022; 20:2722-2732. [PMID: 36074019 PMCID: PMC9826392 DOI: 10.1111/jth.15873] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/29/2022] [Accepted: 09/06/2022] [Indexed: 01/13/2023]
Abstract
Recombinant ADAMTS13 is currently undergoing clinical trials as a treatment for hereditary thrombotic thrombocytopenic purpura, a lethal microvascular condition resulting from ADAMTS13 deficiency. Preclinical studies have also demonstrated its efficacy in treating arterial thrombosis and inflammation without causing bleeding, suggesting that recombinant ADAMTS13 may have broad applicability as an antithrombotic agent. Despite this progress, we currently do not understand the mechanisms that regulate ADAMTS13 activity in vivo. ADAMTS13 evades canonical means of protease regulation because it is secreted as an active enzyme and has a long half-life in circulation, suggesting that it is not inhibited by natural protease inhibitors. Although shear can spatially and temporally activate von Willebrand factor to capture circulating platelets, it is also required for cleavage by ADAMTS13. Therefore, spatial and temporal regulation of ADAMTS13 activity may be required to stabilize von Willebrand factor-platelet strings at sites of vascular injury. This review outlines potential mechanisms that regulate ADAMTS13 in vivo including shear-dependency, local inactivation, and biochemical and structural regulation of substrate binding. Recently published structural data of ADAMTS13 is discussed, which may help to generate novel hypotheses for future research.
Collapse
Affiliation(s)
- Veronica DeYoung
- Department of Medicine, McMaster UniversityThrombosis and Atherosclerosis Research InstituteHamiltonOntarioCanada
| | - Kanwal Singh
- Department of Medicine, McMaster UniversityThrombosis and Atherosclerosis Research InstituteHamiltonOntarioCanada
| | - Colin A. Kretz
- Department of Medicine, McMaster UniversityThrombosis and Atherosclerosis Research InstituteHamiltonOntarioCanada
| |
Collapse
|
15
|
Zhang L, Ma J, Yang F, Li S, Ma W, Chang X, Yang L. Neuroprotective Effects of Quercetin on Ischemic Stroke: A Literature Review. Front Pharmacol 2022; 13:854249. [PMID: 35662707 PMCID: PMC9158527 DOI: 10.3389/fphar.2022.854249] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/28/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke (IS) is characterized by high recurrence and disability; however, its therapies are very limited. As one of the effective methods of treating acute attacks of IS, intravenous thrombolysis has a clear time window. Quercetin, a flavonoid widely found in vegetables and fruits, inhibits immune cells from secreting inflammatory cytokines, thereby reducing platelet aggregation and limiting inflammatory thrombosis. In pre-clinical studies, it has been shown to exhibit neuroprotective effects in patients with ischemic brain injury. However, its specific mechanism of action remains unknown. Therefore, this review aims to use published data to elucidate the potential value of quercetin in patients with ischemic brain injury. This article also reviews the plant sources, pharmacological effects, and metabolic processes of quercetin in vivo, thus focusing on its mechanism in inhibiting immune cell activation and inflammatory thrombosis as well as promoting neuroprotection against ischemic brain injury.
Collapse
Affiliation(s)
- Leilei Zhang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Jingying Ma
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Fan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Sishi Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Wangran Ma
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiang Chang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Lin Yang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| |
Collapse
|
16
|
Girolamo F, Errede M, Bizzoca A, Virgintino D, Ribatti D. Central Nervous System Pericytes Contribute to Health and Disease. Cells 2022; 11:1707. [PMID: 35626743 PMCID: PMC9139243 DOI: 10.3390/cells11101707] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/11/2022] Open
Abstract
Successful neuroprotection is only possible with contemporary microvascular protection. The prevention of disease-induced vascular modifications that accelerate brain damage remains largely elusive. An improved understanding of pericyte (PC) signalling could provide important insight into the function of the neurovascular unit (NVU), and into the injury-provoked responses that modify cell-cell interactions and crosstalk. Due to sharing the same basement membrane with endothelial cells, PCs have a crucial role in the control of endothelial, astrocyte, and oligodendrocyte precursor functions and hence blood-brain barrier stability. Both cerebrovascular and neurodegenerative diseases impair oxygen delivery and functionally impair the NVU. In this review, the role of PCs in central nervous system health and disease is discussed, considering their origin, multipotency, functions and also dysfunction, focusing on new possible avenues to modulate neuroprotection. Dysfunctional PC signalling could also be considered as a potential biomarker of NVU pathology, allowing us to individualize therapeutic interventions, monitor responses, or predict outcomes.
Collapse
Affiliation(s)
- Francesco Girolamo
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Mariella Errede
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Antonella Bizzoca
- Physiology Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Daniela Virgintino
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Domenico Ribatti
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| |
Collapse
|
17
|
Boltze J, Perez-Pinzon MA. Focused Update on Stroke Neuroimmunology: Current Progress in Preclinical and Clinical Research and Recent Mechanistic Insight. Stroke 2022; 53:1432-1437. [PMID: 35467998 DOI: 10.1161/strokeaha.122.039005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Local and systemic inflammation contribute significantly to stroke risk factors as well as determining stroke impact and outcome. Previously being considered as an immuno-privileged domain, the central nervous system is now recognized for multiple and complex interactions with the immune system in health and disease. The sterile inflammatory response emerging after ischemic stroke is a major pathophysiological hallmark and considered to be a promising therapeutic target. Even (mal)adaptive immune responses following stroke, potentially contributing to long-term impact and outcome, are increasingly discussed. However, the complex interaction between the central nervous and the immune system are only partially understood, placing neuroimmunological investigations at the forefront of preclinical and clinical research. This Focused Update summarizes current knowledge in stroke neuroimmunology across all relevant disciplines and discusses major advances as well as recent mechanistic insights. Specifically, neuroimmunological processes and neuroinflammation following ischemic are discussed in the context of blood-brain barrier dysfunction, microglia activation, thromboinflammation, and sex differences in poststroke neuroimmunological responses. The Focused Update further highlights advances in neuroimaging and experimental treatments to visualize and counter neuroinflammatory consequences of ischemic stroke.
Collapse
Affiliation(s)
- Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom (J.B.)
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, FL (M.A.P.-P.)
| |
Collapse
|
18
|
Nayak MK, Flora GD, Chauhan AK. Constitutively active ADAMTS13: An emerging thrombolytic agent for acute ischemic stroke. J Thromb Haemost 2022; 20:790-793. [PMID: 35106938 PMCID: PMC9029329 DOI: 10.1111/jth.15649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Manasa K Nayak
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Gagan D Flora
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Anil K Chauhan
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
19
|
Residues R1075, D1090, R1095, and C1130 Are Critical in ADAMTS13 TSP8-Spacer Interaction Predicted by Molecular Dynamics Simulation. Molecules 2021; 26:molecules26247525. [PMID: 34946607 PMCID: PMC8703438 DOI: 10.3390/molecules26247525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022] Open
Abstract
ADAMTS13 (A Disintegrin and Metalloprotease with Thrombospondin type 1 repeats, member 13) cleaves von Willebrand Factor (VWF) multimers to limit the prothrombotic function of VWF. The deficiency of ADAMTS13 causes a lethal thrombotic microvascular disease, thrombotic thrombocytopenic purpura (TTP). ADAMTS13 circulates in a “closed” conformation with the distal domain associating the Spacer domain to avoid off-target proteolysis or recognition by auto-antibodies. However, the interactions of the distal TSP8 domain and the Spacer domain remain elusive. Here, we constructed the TSP8-Spacer complex by a combination of homology modelling and flexible docking. Molecular dynamics simulation was applied to map the binding sites on the TSP8 or Spacer domain. The results predicted that R1075, D1090, R1095, and C1130 on the TSP8 domain were key residues that interacted with the Spacer domain. R1075 and R1095 bound exosite-4 tightly, D1090 formed multiple hydrogen bonds and salt bridges with exosite-3, and C1130 interacted with both exosite-3 and exosite-4. Specific mutations of exosite-3 (R568K/F592Y/R660K/Y661F/Y665F) or the four key residues (R1075A/D1090A/R1095A/C1130A) impaired the binding of the TSP8 domain to the Spacer domain. These results shed new light on the understanding of the auto-inhibition of ADAMTS13.
Collapse
|