1
|
Roesler J, Spitzer D, Jia X, Aasen SN, Sommer K, Roller B, Olshausen N, Hebach NR, Albinger N, Ullrich E, Zhu L, Wang F, Macas J, Forster MT, Steinbach JP, Sevenich L, Devraj K, Thorsen F, Karreman MA, Plate KH, Reiss Y, Harter PN. Disturbance in cerebral blood microcirculation and hypoxic-ischemic microenvironment are associated with the development of brain metastasis. Neuro Oncol 2024; 26:2084-2099. [PMID: 38831719 PMCID: PMC11534324 DOI: 10.1093/neuonc/noae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Brain metastases (BM) constitute an increasing challenge in oncology due to their impact on neurological function, limited treatment options, and poor prognosis. BM occurs through extravasation of circulating tumor cells across the blood-brain barrier. However, the extravasation processes are still poorly understood. We here propose a brain colonization process which mimics infarction-like microenvironmental reactions, that are dependent on Angiopoietin-2 (Ang-2) and vascular endothelial growth factor (VEGF). METHODS In this study, intracardiac BM models were used, and cerebral blood microcirculation was monitored by 2-photon microscopy through a cranial window. BM formation was observed using cranial magnetic resonance, bioluminescent imaging, and postmortem autopsy. Ang-2/VEGF targeting strategies and Ang-2 gain-of-function (GOF) mice were employed to interfere with BM formation. In addition, vascular and stromal factors as well as clinical outcomes were analyzed in BM patients. RESULTS Blood vessel occlusions by cancer cells were detected, accompanied by significant disturbances of cerebral blood microcirculation, and focal stroke-like histological signs. Cerebral endothelial cells showed an elevated Ang-2 expression both in mouse and human BM. Ang-2 GOF resulted in an increased BM burden. Combined anti-Ang-2/anti-VEGF therapy led to a decrease in brain metastasis size and number. Ang-2 expression in tumor vessels of established human BM negatively correlated with survival. CONCLUSIONS Our observations revealed a relationship between disturbance of cerebral blood microcirculation and brain metastasis formation. This suggests that vessel occlusion by tumor cells facilitates brain metastatic extravasation and seeding, while combined inhibition of microenvironmental effects of Ang-2 and VEGF prevents the outgrowth of macrometastases.
Collapse
Affiliation(s)
- Jenny Roesler
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Daniel Spitzer
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Xiaoxiong Jia
- Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
- Neurosurgery Department, Tianjin Huanhu Hospital, Tianjin, China
| | - Synnøve Nymark Aasen
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway
| | - Kathleen Sommer
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Bastian Roller
- Goethe University, University Hospital, Dr. Senckenberg Institute for Neurooncology, Frankfurt, Germany
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Niels Olshausen
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils R Hebach
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nawid Albinger
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Department of Pediatrics, Experimental Immunology and Cell Therapy, Goethe University, University Hospital, Frankfurt, Germany
| | - Evelyn Ullrich
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Department of Pediatrics, Experimental Immunology and Cell Therapy, Goethe University, University Hospital, Frankfurt, Germany
| | - Ling Zhu
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Fan Wang
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Jadranka Macas
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Marie-Therese Forster
- Department of Neurosurgery, Goethe University, University Hospital, Frankfurt, Germany
| | - Joachim P Steinbach
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, University Hospital, Dr. Senckenberg Institute for Neurooncology, Frankfurt, Germany
| | - Lisa Sevenich
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Frankfurt, Germany
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
| | - Kavi Devraj
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Hyderabad, India
| | - Frits Thorsen
- Department of Biomedicine, Molecular Imaging Center, University of Bergen, Bergen, Norway
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China
- Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway
| | - Matthia A Karreman
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl H Plate
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Yvonne Reiss
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| | - Patrick N Harter
- Center for Neuropathology and Prion Research, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University, University Hospital, Institute of Neurology (Edinger Institute), Frankfurt, Germany
| |
Collapse
|
2
|
Wang J, Jia R, Wan W, Han H, Wang G, Li Z, Li J. Drug Delivery Targeting Neuroinflammation to Treat Brain Diseases. Bioconjug Chem 2024. [PMID: 39377704 DOI: 10.1021/acs.bioconjchem.4c00414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Inflammation within the brain is a hallmark of a wide range of brain diseases. The complex role of inflammatory processes in these conditions suggests that neuroinflammation could be a valuable therapeutic target. While several promising anti-inflammatory agents have been identified, their clinical application in brain diseases is often hampered by the inability to cross the blood-brain barrier (BBB) and reach therapeutically effective concentrations at the pathological sites. This limitation highlights the urgent need for effective BBB-penetrating drug delivery systems designed to target brain inflammation. This review critically examines the recent advances over the past five years in drug delivery strategies aimed at mitigating brain inflammation in Alzheimer's disease and ischemic stroke─two of the leading causes of death and disability worldwide. Additionally, we address the key challenges in this field, offering insights into future directions for targeting neuroinflammation in the treatment of brain diseases.
Collapse
Affiliation(s)
- Juntao Wang
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100, China
| | - Ruiqin Jia
- School of Pharmacy, Henan University, Kaifeng, 475001, China
| | - Wubo Wan
- Yazhou Bay Innovation Institute, Hainan Tropical Ocean University, Sanya, 572022, China
| | - Haijun Han
- Yazhou Bay Innovation Institute, Hainan Tropical Ocean University, Sanya, 572022, China
| | - Guoying Wang
- Macquarie Medical School, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Zhen Li
- Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Jia Li
- Macquarie Medical School, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
3
|
Sha L, Zhao Y, Li S, Wei D, Tao Y, Wang Y. Insights to Ang/Tie signaling pathway: another rosy dawn for treating retinal and choroidal vascular diseases. J Transl Med 2024; 22:898. [PMID: 39367441 PMCID: PMC11451039 DOI: 10.1186/s12967-024-05441-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/27/2024] [Indexed: 10/06/2024] Open
Abstract
Retinal neurovascular unit (NVU) is a multi-cellular structure that consists of the functional coupling between neural tissue and vascular system. Disrupted NVU will result in the occurrence of retinal and choroidal vascular diseases, which are characterized by the development of neovascularization, increased vascular permeability, and inflammation. This pathological entity mainly includes neovascular age-related macular degeneration (neovascular-AMD), diabetic retinopathy (DR) retinal vein occlusion (RVO), and retinopathy of prematurity (ROP). Emerging evidences suggest that the angopoietin/tyrosine kinase with immunoglobulin and epidermal growth factor homology domains (Ang/Tie) signaling pathway is essential for the development of retinal and choroidal vascular. Tie receptors and their downstream pathways play a key role in modulating the vascular development, vascular stability, remodeling and angiogenesis. Angiopoietin 1 (Ang1) is a natural agonist of Tie2 receptor, which can promote vascular stability. On the other hand, angiopoietin 2 (Ang2) is an antagonist of Tie2 receptor that causes vascular instability. Currently, agents targeting the Ang/Tie signaling pathway have been used to inhibit neovascularization and vascular leakage in neovascular-AMD and DR animal models. Particularly, the AKB-9778 and Faricimab have shown promising efficacy in improving visual acuity in patients with neovascular-AMD and DR. These experimental and clinical evidences suggest that activation of Ang/Tie signaling pathway can inhibit the vascular permeability, neovascularization, thereby maintaining the normal function and structure of NVU. This review seeks to introduce the versatile functions and elucidate the modulatory mechanisms of Ang/Tie signaling pathway. Recent pharmacologic therapies targeting this pathway are also elaborated and summarized. Further translation of these findings may afford a new therapeutic strategy from bench to bedside.
Collapse
Affiliation(s)
- Lulu Sha
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yameng Zhao
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Dong Wei
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Ye Tao
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| | - Yange Wang
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
4
|
Alvarez-Olmedo D, Kamaliddin C, Verhey TB, Ho M, DeVinney R, Chaconas G. Transendothelial migration of the Lyme disease spirochete involves spirochete internalization as an intermediate step through a transcellular pathway that involves Cdc42 and Rac1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612329. [PMID: 39314306 PMCID: PMC11419014 DOI: 10.1101/2024.09.10.612329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Despite its importance in pathogenesis, the hematogenous dissemination pathway of B. burgdorferi is still largely uncharacterized. To probe the molecular details of transendothelial migration more easily, we studied this process using cultured primary or telomerase-immortalized human microvascular endothelial cells in a medium that maintains both the human cells and the spirochetes. In B. burgdorferi infected monolayers we observed ∼55% of wild-type spirochetes crossing the monolayer. Microscopic characterization revealed entrance points across the cellular surface rather than at cellular junctions, supporting a transcellular route. In support of this pathway, locking the endothelial junctions using a VE-PTP inhibitor did not reduce transendothelial migration. We also used inhibitors to block the most common endocytic pathways to elucidate effectors that might be involved in B. burgdorferi uptake and/or transmigration. Directly inhibiting Cdc42 reduced spirochete transmigration by impeding internalization. However, blocking Rac1 alone dramatically reduced transmigration and resulted in a concomitant increase in spirochete accumulation in the cell. Our combined results support that B. burgdorferi internalization is an intermediate step in the transendothelial migration process which requires both Cdc42 and Rac1; Cdc42 is needed for spirochete internalization while Rac1 is required for cellular egress. These are the first two host proteins implicated in B. burgdorferi transmigration across endothelial cells. IMPORTANCE Lyme borreliosis is caused by Borrelia burgdorferi and related bacteria. It is the most common tick-transmitted illness in the Northern Hemisphere. The ability of this pathogen to spread to a wide variety of locations results in a diverse set of clinical manisfestations, yet little is known regarding vascular escape of the spirochete, an important pathway for dissemination. Our current work has studied the traversal of B. burgdorferi across a monolayer of microvascular endothelial cells grown in culture. We show that this occurs by passage of the spirochetes directly through these cells rather than at cellular junctions and that internalization of B. burgdorferi is an intermediate step in the transmigration process. We also identify the first two host proteins, Cdc42 and Rac1, that are used by the spirochetes to promote traversal of the cellular monolayer. Our new experimental system also provides a new avenue for further studies of this important process.
Collapse
|
5
|
Dave JM, Chakraborty R, Agyemang A, Ntokou A, Saito J, Ballabh P, Martin KA, Greif DM. Loss of TGFβ-Mediated Repression of Angiopoietin-2 in Pericytes Underlies Germinal Matrix Hemorrhage Pathogenesis. Stroke 2024; 55:2340-2352. [PMID: 39129597 PMCID: PMC11347087 DOI: 10.1161/strokeaha.123.045248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND TGF (transforming growth factor)-β pathway is central to blood-brain barrier development as it regulates cross talk between pericytes and endothelial cells. Murine embryos lacking TGFβ receptor Alk5 (activin receptor-like kinase 5) in brain pericytes (mutants) display endothelial cell hyperproliferation, abnormal vessel morphology, and gross germinal matrix hemorrhage-intraventricular hemorrhage (GMH-IVH), leading to perinatal lethality. Mechanisms underlying how ALK5 signaling in pericytes noncell autonomously regulates endothelial cell behavior remain elusive. METHODS Transcriptomic analysis of human brain pericytes with ALK5 silencing identified differential gene expression. Brain vascular cells isolated from mutant embryonic mice with GMH-IVH and preterm human IVH brain samples were utilized for target validation. Finally, pharmacological and genetic inhibition was used to study the therapeutic effects on GMH-IVH pathology. RESULTS Herein, we establish that the TGFβ/ALK5 pathway robustly represses ANGPT2 (angiopoietin-2) in pericytes via epigenetic remodeling. TGFβ-driven SMAD (suppressor of mothers against decapentaplegic) 3/4 associates with TGIF1 (TGFβ-induced factor homeobox 1) and HDAC (histone deacetylase) 5 to form a corepressor complex at the Angpt2 promoter, resulting in promoter deacetylation and gene repression. Moreover, murine and human germinal matrix vessels display increased ANGPT2 expression during GMH-IVH. Isolation of vascular cells from murine germinal matrix identifies pericytes as a cellular source of excessive ANGPT2. In addition, mutant endothelial cells exhibit higher phosphorylated TIE2 (tyrosine protein kinase receptor). Pharmacological or genetic inhibition of ANGPT2 in mutants improves germinal matrix vessel morphology and attenuates GMH pathogenesis. Importantly, genetic ablation of Angpt2 in mutant pericytes prevents perinatal lethality, prolonging survival. CONCLUSIONS This study demonstrates that TGFβ-mediated ANGPT2 repression in pericytes is critical for maintaining blood-brain barrier integrity and identifies pericyte-derived ANGPT2 as an important pathological target for GMH-IVH.
Collapse
Affiliation(s)
- Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Raja Chakraborty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Alex Agyemang
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aglaia Ntokou
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathleen A. Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
6
|
Kuriyama A, Nakamura S, Inokuchi Y, Abe H, Yasuda H, Hidaka Y, Nagaoka K, Soeda T, Shimazawa M, Hara H. The protective effect of anti-VEGF-A/Ang-2 bispecific antibody on retinal vein occlusion model mice. Eur J Pharmacol 2024; 976:176691. [PMID: 38821166 DOI: 10.1016/j.ejphar.2024.176691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
(233/250) Retinal vein occlusion (RVO) causes macular edema and retinal ischemia resulting in visual field and vision loss. A bispecific antibody that blocks VEGF-A and angiopoietin-2 (Ang-2) has been recently launched and applied clinically to treat macular edema, but the role of Ang-2 in the pathogenesis of RVO is still unclear. In this study, we investigated the effects of the anti-VEGF-A/anti-Ang-2 bispecific antibody (BsAb) in a murine RVO model. By using RVO model mice, the expression of Ang-2 gene and protein was examined in the retina through real-time qPCR and Western blotting, respectively. A significant increase in Ang-2 was detected 1 day after occlusion. Immediately after occlusion, control IgG 400 μg/mL, anti-VEGF-A antibody 200 μg/mL, anti-Ang-2 antibody 200 μg/mL, and BsAb 400 μg/mL were intravitreally administered at 2 μL. Visual function was examined using electroretinograms, and apoptosis was examined using TUNEL staining. Interestingly, BsAb partially suppressed the decrease in amplitude of a and b waves compared to control IgG. Anti-Ang-2 antibody and BsAb reduced apoptosis-positive cells 1 day after occlusion. Comprehensive gene expression profiles were also examined using RNA sequencing analysis. RNA sequencing analysis of the retinal tissues showed that BsAb suppressed expression of gene groups associated with inflammatory response and vascular development compared to anti-VEGF-A antibody. Taken together, higher expression of Ang-2 contributes to the pathophysiology of RVO, providing a possible mechanism for the efficacy of BsAb in suppressing retinal dysfunction in RVO.
Collapse
Affiliation(s)
- Aika Kuriyama
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuta Inokuchi
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Hiroto Abe
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Hiroto Yasuda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yae Hidaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Koki Nagaoka
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Tetsuhiro Soeda
- Product Research Dept., Chugai Pharmaceutical Co., Ltd. Kanagawa, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
7
|
Cen K, Huang Y, Xie Y, Liu Y. The guardian of intracranial vessels: Why the pericyte? Biomed Pharmacother 2024; 176:116870. [PMID: 38850658 DOI: 10.1016/j.biopha.2024.116870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Intracranial atherosclerotic stenosis (ICAS) is a pathological condition characterized by progressive narrowing or complete blockage of intracranial blood vessels caused by plaque formation. This condition leads to reduced blood flow to the brain, resulting in cerebral ischemia and hypoxia. Ischemic stroke (IS) resulting from ICAS poses a significant global public health challenge, especially among East Asian populations. However, the underlying causes of the notable variations in prevalence among diverse populations, as well as the most effective strategies for preventing and treating the rupture and blockage of intracranial plaques, remain incompletely comprehended. Rupture of plaques, bleeding, and thrombosis serve as precipitating factors in the pathogenesis of luminal obstruction in intracranial arteries. Pericytes play a crucial role in the structure and function of blood vessels and face significant challenges in regulating the Vasa Vasorum (VV)and preventing intraplaque hemorrhage (IPH). This review aims to explore innovative therapeutic strategies that target the pathophysiological mechanisms of vulnerable plaques by modulating pericyte biological function. It also discusses the potential applications of pericytes in central nervous system (CNS) diseases and their prospects as a therapeutic intervention in the field of biological tissue engineering regeneration.
Collapse
Affiliation(s)
- Kuan Cen
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YinFei Huang
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - Yu Xie
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YuMin Liu
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China.
| |
Collapse
|
8
|
Sun P, Yang Y, Yang L, Qian Y, Liang M, Chen H, Zhang J, Qiu Y, Guo L, Fu S. Quercetin Protects Blood-Brain Barrier Integrity via the PI3K/Akt/Erk Signaling Pathway in a Mouse Model of Meningitis Induced by Glaesserella parasuis. Biomolecules 2024; 14:696. [PMID: 38927100 PMCID: PMC11201931 DOI: 10.3390/biom14060696] [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: 05/08/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Glaesserella parasuis (G. parasuis) causes serious inflammation and meningitis in piglets. Quercetin has anti-inflammatory and anti-bacterial activities; however, whether quercetin can alleviate brain inflammation and provide protective effects during G. parasuis infection has not been studied. Here, we established a mouse model of G. parasuis infection in vivo and in vitro to investigate transcriptome changes in the mouse cerebrum and determine the protective effects of quercetin on brain inflammation and blood-brain barrier (BBB) integrity during G. parasuis infection. The results showed that G. parasuis induced brain inflammation, destroyed BBB integrity, and suppressed PI3K/Akt/Erk signaling-pathway activation in mice. Quercetin decreased the expression of inflammatory cytokines (Il-18, Il-6, Il-8, and Tnf-α) and BBB-permeability marker genes (Mmp9, Vegf, Ang-2, and Et-1), increased the expression of angiogenetic genes (Sema4D and PlexinB1), reduced G. parasuis-induced tight junction disruption, and reactivated G. parasuis-induced suppression of the PI3K/Akt/Erk signaling pathway in vitro. Thus, we concluded that quercetin may protect BBB integrity via the PI3K/Akt/Erk signaling pathway during G. parasuis infection. This was the first attempt to explore the protective effects of quercetin on brain inflammation and BBB integrity in a G. parasuis-infected mouse model. Our findings indicated that quercetin is a promising natural agent for the prevention and treatment of G. parasuis infection.
Collapse
Affiliation(s)
- Peiyan Sun
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yaqiong Yang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Linrong Yang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuanzhuo Qian
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mingxia Liang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Hongbo Chen
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan 430023, China
| | - Jing Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan 430023, China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Ling Guo
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| |
Collapse
|
9
|
Mohammadi H, Ariaei A, Ghobadi Z, Gorgich EAC, Rustamzadeh A. Which neuroimaging and fluid biomarkers method is better in theranostic of Alzheimer's disease? An umbrella review. IBRO Neurosci Rep 2024; 16:403-417. [PMID: 38497046 PMCID: PMC10940808 DOI: 10.1016/j.ibneur.2024.02.007] [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: 12/11/2023] [Accepted: 02/24/2024] [Indexed: 03/19/2024] Open
Abstract
Biomarkers are measured to evaluate physiological and pathological processes as well as responses to a therapeutic intervention. Biomarkers can be classified as diagnostic, prognostic, predictor, clinical, and therapeutic. In Alzheimer's disease (AD), multiple biomarkers have been reported so far. Nevertheless, finding a specific biomarker in AD remains a major challenge. Three databases, including PubMed, Web of Science, and Scopus were selected with the keywords of Alzheimer's disease, neuroimaging, biomarker, and blood. The results were finalized with 49 potential CSF/blood and 35 neuroimaging biomarkers. To distinguish normal from AD patients, amyloid-beta42 (Aβ42), plasma glial fibrillary acidic protein (GFAP), and neurofilament light (NFL) as potential biomarkers in cerebrospinal fluid (CSF) as well as the serum could be detected. Nevertheless, most of the biomarkers fairly change in the CSF during AD, listed as kallikrein 6, virus-like particles (VLP-1), galectin-3 (Gal-3), and synaptotagmin-1 (Syt-1). From the neuroimaging aspect, atrophy is an accepted biomarker for the neuropathologic progression of AD. In addition, Magnetic resonance spectroscopy (MRS), diffusion weighted imaging (DWI), diffusion tensor imaging (DTI), tractography (DTT), positron emission tomography (PET), and functional magnetic resonance imaging (fMRI), can be used to detect AD. Using neuroimaging and CSF/blood biomarkers, in combination with artificial intelligence, it is possible to obtain information on prognosis and follow-up on the different stages of AD. Hence physicians could select the suitable therapy to attenuate disease symptoms and follow up on the efficiency of the prescribed drug.
Collapse
Affiliation(s)
- Hossein Mohammadi
- Department of Bioimaging, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences (MUI), Isfahan, Islamic Republic of Iran
| | - Armin Ariaei
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Zahra Ghobadi
- Advanced Medical Imaging Ward, Pars Darman Medical Imaging Center, Karaj, Islamic Republic of Iran
| | - Enam Alhagh Charkhat Gorgich
- Department of Anatomy, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Islamic Republic of Iran
| | - Auob Rustamzadeh
- Cellular and Molecular Research Center, Research Institute for Non-communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
10
|
Baedorf-Kassis E, Murn M, Dzierba AL, Serra AL, Garcia I, Minus E, Padilla C, Sarge T, Goodspeed VM, Matthay MA, Gong MN, Cook D, Loring SH, Talmor D, Beitler JR. Respiratory drive heterogeneity associated with systemic inflammation and vascular permeability in acute respiratory distress syndrome. Crit Care 2024; 28:136. [PMID: 38654391 PMCID: PMC11036740 DOI: 10.1186/s13054-024-04920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND In acute respiratory distress syndrome (ARDS), respiratory drive often differs among patients with similar clinical characteristics. Readily observable factors like acid-base state, oxygenation, mechanics, and sedation depth do not fully explain drive heterogeneity. This study evaluated the relationship of systemic inflammation and vascular permeability markers with respiratory drive and clinical outcomes in ARDS. METHODS ARDS patients enrolled in the multicenter EPVent-2 trial with requisite data and plasma biomarkers were included. Neuromuscular blockade recipients were excluded. Respiratory drive was measured as PES0.1, the change in esophageal pressure during the first 0.1 s of inspiratory effort. Plasma angiopoietin-2, interleukin-6, and interleukin-8 were measured concomitantly, and 60-day clinical outcomes evaluated. RESULTS 54.8% of 124 included patients had detectable respiratory drive (PES0.1 range of 0-5.1 cm H2O). Angiopoietin-2 and interleukin-8, but not interleukin-6, were associated with respiratory drive independently of acid-base, oxygenation, respiratory mechanics, and sedation depth. Sedation depth was not significantly associated with PES0.1 in an unadjusted model, or after adjusting for mechanics and chemoreceptor input. However, upon adding angiopoietin-2, interleukin-6, or interleukin-8 to models, lighter sedation was significantly associated with higher PES0.1. Risk of death was less with moderate drive (PES0.1 of 0.5-2.9 cm H2O) compared to either lower drive (hazard ratio 1.58, 95% CI 0.82-3.05) or higher drive (2.63, 95% CI 1.21-5.70) (p = 0.049). CONCLUSIONS Among patients with ARDS, systemic inflammatory and vascular permeability markers were independently associated with higher respiratory drive. The heterogeneous response of respiratory drive to varying sedation depth may be explained in part by differences in inflammation and vascular permeability.
Collapse
Affiliation(s)
- Elias Baedorf-Kassis
- Division of Pulmonary and Critical Care Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael Murn
- Columbia Respiratory Critical Care Trials Group, Columbia University College of Physicians and Surgeons, and New York-Presbyterian Hospital, 622 West 168th Street, New York, NY, 10032, USA
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, NY, USA
| | - Amy L Dzierba
- Columbia Respiratory Critical Care Trials Group, Columbia University College of Physicians and Surgeons, and New York-Presbyterian Hospital, 622 West 168th Street, New York, NY, 10032, USA
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, NY, USA
- Department of Pharmacy, New York-Presbyterian Hospital, New York, NY, USA
| | - Alexis L Serra
- Columbia Respiratory Critical Care Trials Group, Columbia University College of Physicians and Surgeons, and New York-Presbyterian Hospital, 622 West 168th Street, New York, NY, 10032, USA
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, NY, USA
| | - Ivan Garcia
- Columbia Respiratory Critical Care Trials Group, Columbia University College of Physicians and Surgeons, and New York-Presbyterian Hospital, 622 West 168th Street, New York, NY, 10032, USA
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, NY, USA
| | - Emily Minus
- Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | - Clarissa Padilla
- Columbia Respiratory Critical Care Trials Group, Columbia University College of Physicians and Surgeons, and New York-Presbyterian Hospital, 622 West 168th Street, New York, NY, 10032, USA
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, NY, USA
| | - Todd Sarge
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Valerie M Goodspeed
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | - Michelle N Gong
- Department of Critical Care Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deborah Cook
- St. Joseph's Hospital and McMaster University, Hamilton, ON, Canada
| | - Stephen H Loring
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel Talmor
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jeremy R Beitler
- Columbia Respiratory Critical Care Trials Group, Columbia University College of Physicians and Surgeons, and New York-Presbyterian Hospital, 622 West 168th Street, New York, NY, 10032, USA.
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
11
|
Pociūtė A, Pivoriūnas A, Verkhratsky A. Astrocytes dynamically regulate the blood-brain barrier in the healthy brain. Neural Regen Res 2024; 19:709-710. [PMID: 37843196 PMCID: PMC10664108 DOI: 10.4103/1673-5374.382248] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/27/2023] [Accepted: 07/14/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
- Agnė Pociūtė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Alexei Verkhratsky
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
12
|
Jia L, Li X, Shen J, Teng Y, Zhang B, Zhang M, Gu Y, Xu H. Ang-1, Ang-2, and Tie2 are diagnostic biomarkers for Henoch-Schönlein purpura and pediatric-onset systemic lupus erythematous. Open Life Sci 2024; 19:20220812. [PMID: 38465338 PMCID: PMC10921503 DOI: 10.1515/biol-2022-0812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 03/12/2024] Open
Abstract
Henoch-Schönlein purpura (HSP) and pediatric-onset systemic lupus erythematosus (pSLE) are closely associated with vasculitis and vascular diseases. This study aimed to investigate the clinical diagnostic values of Ang-1, Ang-2, and Tie2 for HSP and pSLE. We surveyed 82 HSP patients, 34 pSLE patients, and 10 healthy children. The expression levels of Ang-1, Ang-2, and Tie2 in the serum and urine were assessed using enzyme-linked immunosorbent assay. The diagnostic values of Ang-1, Ang-2, and Tie2 for HSP and pSLE were evaluated using receiver operating characteristic curve analysis. The results revealed that the serum and urine expression levels of Ang-2 and Tie2 were significantly elevated in HSP and pSLE patients, whereas the Ang-1/Ang-2 values were reduced. Additionally, Ang-1 was highly expressed in the serum and urine of HSP patients and in the serum of pSLE patients. Ang-1, Ang-2, and Tie2 showed differential expression in various types of HSP and pSLE compared with their expression in healthy controls. In summary, Ang-1, Ang-2, and Tie2 can serve as biomarkers for HSP and pSLE. Moreover, Ang-1/Ang-2 values are reduced in HSP and pSLE patients. Ang-1, Ang-2, and Tie2 can be used as biomarkers for HSP and pSLE.
Collapse
Affiliation(s)
- Lishan Jia
- Department of Pediatrics, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, No. 58 Changsheng South Road, Taicang City, Jiangsu Province, 215400, China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children’s Hospital of Soochow University, No. 303 Jingde Road, Gusu District, Suzhou City, Jiangsu Province, 215003, China
| | - Jiayun Shen
- Department of Pediatrics, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, No. 58 Changsheng South Road, Taicang City, Jiangsu Province, 215400, China
| | - Yan Teng
- Department of Pediatrics, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, No. 58 Changsheng South Road, Taicang City, Jiangsu Province, 215400, China
| | - Baoqin Zhang
- Department of Pediatrics, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, No. 58 Changsheng South Road, Taicang City, Jiangsu Province, 215400, China
| | - Min Zhang
- Department of Pediatrics, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, No. 58 Changsheng South Road, Taicang City, Jiangsu Province, 215400, China
| | - Yueqin Gu
- Department of Pediatrics, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, No. 58 Changsheng South Road, Taicang City, Jiangsu Province, 215400, China
| | - Hong Xu
- Department of Nephrology, Children’s Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai City, 201102, China
| |
Collapse
|
13
|
Van Hulle C, Ince S, Okonkwo OC, Bendlin BB, Johnson SC, Carlsson CM, Asthana S, Love S, Blennow K, Zetterberg H, Scott Miners J. Elevated CSF angiopoietin-2 correlates with blood-brain barrier leakiness and markers of neuronal injury in early Alzheimer's disease. Transl Psychiatry 2024; 14:3. [PMID: 38182581 PMCID: PMC10770135 DOI: 10.1038/s41398-023-02706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/12/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Breakdown of the neurovascular unit is associated with blood-brain barrier (BBB) leakiness contributing to cognitive decline and disease pathology in the early stages of Alzheimer's disease (AD). Vascular stability depends on angiopoietin-1 (ANGPT-1) signalling, antagonised by angiopoietin-2 (ANGPT-2) expressed upon endothelial injury. We examined the relationship between CSF ANGPT-2 and CSF markers of BBB leakiness and core AD biomarkers across three independent cohorts: (i) 31 AD patients and 33 healthy controls grouped according to their biomarker profile (i.e., AD cases t-tau > 400 pg/mL, p-tau > 60 pg/mL and Aβ42 < 550 pg/mL); (ii) 121 participants in the Wisconsin Registry for Alzheimer's Prevention or Wisconsin Alzheimer's Disease Research study (84 participants cognitively unimpaired (CU) enriched for a parental history of AD, 20 participants with mild cognitive impairment (MCI), and 17 with AD); (iii) a neurologically normal cohort aged 23-78 years with paired CSF and serum samples. CSF ANGPT-2, sPDGFRβ, albumin and fibrinogen levels were measured by sandwich ELISA. In cohort (i), CSF ANGPT-2 was elevated in AD and correlated with CSF t-tau and p-tau181 but not Aβ42. ANGPT-2 also correlated positively with CSF sPDGFRβ and fibrinogen - markers of pericyte injury and BBB leakiness. In cohort (ii), CSF ANGPT-2 was highest in MCI and correlated with CSF albumin in the CU and MCI cohorts but not in AD. CSF ANGPT-2 also correlated with CSF t-tau and p-tau and with markers of neuronal injury (neurogranin and α-synuclein) and neuroinflammation (GFAP and YKL-40). In cohort (iii), CSF ANGPT-2 correlated strongly with the CSF/serum albumin ratio. Serum ANGPT-2 showed non-significant positive associations with CSF ANGPT-2 and the CSF/serum albumin ratio. Together, these data indicate that CSF and possibly serum ANGPT-2 is associated with BBB leakiness in early AD and is closely related to tau pathology and neuronal injury. The utility of serum ANGPT-2 as a biomarker of BBB damage in AD requires further study.
Collapse
Affiliation(s)
- Carol Van Hulle
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
| | - Selvi Ince
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Seth Love
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - J Scott Miners
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
14
|
Chi Y, Yu S, Yin J, Liu D, Zhuo M, Li X. Role of Angiopoietin/Tie2 System in Sepsis: A Potential Therapeutic Target. Clin Appl Thromb Hemost 2024; 30:10760296241238010. [PMID: 38449088 PMCID: PMC10921858 DOI: 10.1177/10760296241238010] [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: 01/16/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Sepsis is a disorder of host response caused by severe infection that can lead to life-threatening organ dysfunction. There is no specific treatment for sepsis. Although there are many different pathogens that can cause sepsis, endothelial dysfunction is a frequent mechanism resulting in vascular leakage and coagulation problem. Recent studies on the regulatory pathways of vascular endothelium have shown that the disturbance of angiopoietin (Ang) /Tie2 axis can induce endothelial cell activation, which is the core pathogenesis of sepsis. In this review, we aim to discuss the regulation of Ang/Tie2 axis and the biomarkers involved in the context of sepsis. Also, we attempt to explore the prospective and feasibility of Ang/Tie2 axis as a potential target for sepsis intervention to improve clinical outcomes.
Collapse
Affiliation(s)
- Yawen Chi
- Department of Critical Care Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Sihan Yu
- Department of Critical Care Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jia Yin
- Department of Critical Care Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Danyan Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mengke Zhuo
- Department of Critical Care Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xu Li
- Department of Critical Care Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
15
|
Koltsov IA, Shchukin IA, Fidler MS, Yasamanova AN, Aryasova IK, Boiko AN. [Posterior reversible encephalopathy syndrome in autoimmune disorders]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:50-57. [PMID: 39175240 DOI: 10.17116/jnevro202412407250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Posterior reversible encephalopathy syndrome (PRES) is characterized by nonspecific symptoms, including not only pronounced non-focal and various focal neurological signs but also specific neuroimaging features, including vasogenic edema affecting predominantly the posterior area. PRES usually develops in the setting of acute arterial hypertension. However, it is not uncommon for PRES to develop in non-hypertensive patients, including people with autoimmune disorders (multiple sclerosis, neuromyelitis optica spectrum disorder, etc). PRES could also be due to the toxic effects of drugs or other substances. The pathophysiological mechanisms of PRES include impaired autoregulation of cerebral blood flow due to acute arterial hypertension and toxic endotheliotropic effects of endogenous and exogenous factors.
Collapse
Affiliation(s)
- I A Koltsov
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - I A Shchukin
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - M S Fidler
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A N Yasamanova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - I K Aryasova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A N Boiko
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| |
Collapse
|
16
|
Hu S, Yang B, Shu S, He X, Sang H, Fan X, Zhang H. Targeting Pericytes for Functional Recovery in Ischemic Stroke. Neuromolecular Med 2023; 25:457-470. [PMID: 37166748 DOI: 10.1007/s12017-023-08748-z] [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: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Pericytes surrounding endothelial cells in the capillaries are emerging as an attractive cell resource, which can show a large variety of functions in ischemic stroke, including preservation of the blood-brain barrier, regulation of immune function, and support for cerebral vasculature. These functions have been fully elucidated in previous studies. However, in recent years, increasing evidence has shown that pericytes play an important role in neurological recovery after ischemic stroke due to their regenerative function which can be summarized in two aspects according to current discoveries, one is that pericytes are thought to be multipotential themselves, and the other is that pericytes can promote the differentiation of oligodendrocyte progenitor cells (OPCs). Considering the neuroprotective treatment for stroke has not been much progressed in recent years, new therapies targeting pericytes may be a future direction. Here, we will review the beneficial effects of pericytes in ischemic stroke from two directions: the barrier and vascular functions and the regenerative functions of pericytes.
Collapse
Affiliation(s)
- Shuqi Hu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xudong He
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hongfei Sang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xuemei Fan
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
17
|
Wu W, Huang J, Han P, Zhang J, Wang Y, Jin F, Zhou Y. Research Progress on Natural Plant Molecules in Regulating the Blood-Brain Barrier in Alzheimer's Disease. Molecules 2023; 28:7631. [PMID: 38005352 PMCID: PMC10674591 DOI: 10.3390/molecules28227631] [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: 10/15/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder. With the aging population and the continuous development of risk factors associated with AD, it will impose a significant burden on individuals, families, and society. Currently, commonly used therapeutic drugs such as Cholinesterase inhibitors, N-methyl-D-aspartate antagonists, and multiple AD pathology removal drugs have been shown to have beneficial effects on certain pathological conditions of AD. However, their clinical efficacy is minimal and they are associated with certain adverse reactions. Furthermore, the underlying pathological mechanism of AD remains unclear, posing a challenge for drug development. In contrast, natural plant molecules, widely available, offer multiple targeting pathways and demonstrate inherent advantages in modifying the typical pathologic features of AD by influencing the blood-brain barrier (BBB). We provide a comprehensive review of recent in vivo and in vitro studies on natural plant molecules that impact the BBB in the treatment of AD. Additionally, we analyze their specific mechanisms to offer novel insights for the development of safe and effective targeted drugs as well as guidance for experimental research and the clinical application of drugs for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Weidong Wu
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Jiahao Huang
- Department of Chinese Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Pengfei Han
- Science and Education Section, Zhangjiakou First Hospital, Zhangjiakou 075041, China;
| | - Jian Zhang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Yuxin Wang
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| | - Fangfang Jin
- Department of Internal Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yanyan Zhou
- Basic Theory of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (W.W.); (J.Z.); (Y.W.)
| |
Collapse
|
18
|
Cash A, de Jager C, Brickler T, Soliman E, Ladner L, Kaloss AM, Zhu Y, Pridham KJ, Mills J, Ju J, Basso EKG, Chen M, Johnson Z, Sotiropoulos Y, Wang X, Xie H, Matson JB, Marvin EA, Theus MH. Endothelial deletion of EPH receptor A4 alters single-cell profile and Tie2/Akap12 signaling to preserve blood-brain barrier integrity. Proc Natl Acad Sci U S A 2023; 120:e2204700120. [PMID: 37796990 PMCID: PMC10576133 DOI: 10.1073/pnas.2204700120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 09/06/2023] [Indexed: 10/07/2023] Open
Abstract
Neurobiological consequences of traumatic brain injury (TBI) result from a complex interplay of secondary injury responses and sequela that mediates chronic disability. Endothelial cells are important regulators of the cerebrovascular response to TBI. Our work demonstrates that genetic deletion of endothelial cell (EC)-specific EPH receptor A4 (EphA4) using conditional EphA4f/f/Tie2-Cre and EphA4f/f/VE-Cadherin-CreERT2 knockout (KO) mice promotes blood-brain barrier (BBB) integrity and tissue protection, which correlates with improved motor function and cerebral blood flow recovery following controlled cortical impact (CCI) injury. scRNAseq of capillary-derived KO ECs showed increased differential gene expression of BBB-related junctional and actin cytoskeletal regulators, namely, A-kinase anchor protein 12, Akap12, whose presence at Tie2 clustering domains is enhanced in KO microvessels. Transcript and protein analysis of CCI-injured whole cortical tissue or cortical-derived ECs suggests that EphA4 limits the expression of Cldn5, Akt, and Akap12 and promotes Ang2. Blocking Tie2 using sTie2-Fc attenuated protection and reversed Akap12 mRNA and protein levels cortical-derived ECs. Direct stimulation of Tie2 using Vasculotide, angiopoietin-1 memetic peptide, phenocopied the neuroprotection. Finally, we report a noteworthy rise in soluble Ang2 in the sera of individuals with acute TBI, highlighting its promising role as a vascular biomarker for early detection of BBB disruption. These findings describe a contribution of the axon guidance molecule, EphA4, in mediating TBI microvascular dysfunction through negative regulation of Tie2/Akap12 signaling.
Collapse
Affiliation(s)
- Alison Cash
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, VA24061
| | - Thomas Brickler
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Liliana Ladner
- Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA24016
| | - Alexandra M. Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Yumeng Zhu
- Department of Chemistry, Virginia Tech, Blacksburg, VA24061
| | - Kevin J. Pridham
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Jatia Mills
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Jing Ju
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | | | - Michael Chen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Zachary Johnson
- Genetics, Bioinformatics and Computational Biology Program, Virginia Tech, Blacksburg, VA24061
- Epigenomics and Computational Biology Lab, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA24061
| | - Yianni Sotiropoulos
- Summer Veterinary Student Research Program, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA24061
| | - Xia Wang
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
| | - Hehuang Xie
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
- Genetics, Bioinformatics and Computational Biology Program, Virginia Tech, Blacksburg, VA24061
- Epigenomics and Computational Biology Lab, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA24061
- Center for Engineered Health, Virginia Tech, Blacksburg, VA24061
| | - John B. Matson
- Department of Chemistry, Virginia Tech, Blacksburg, VA24061
| | - Eric A. Marvin
- Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA24016
| | - Michelle H. Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA24061
- Summer Veterinary Student Research Program, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA24061
| |
Collapse
|
19
|
Asano W, Yamanaka K, Ohara Y, Uhara T, Doi S, Orita T, Iwanaga T, Adachi T, Fujioka S, Akaki T, Ikegashira K, Hantani Y. Fragment-Based Discovery of Novel VE-PTP Inhibitors Using Orthogonal Biophysical Techniques. Biochemistry 2023. [PMID: 37414577 DOI: 10.1021/acs.biochem.3c00079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Tyrosine phosphorylation is an essential post-translational modification that regulates various biological events and is implicated in many diseases including cancer and atherosclerosis. Vascular endothelial protein tyrosine phosphatase (VE-PTP), which plays an important role in vascular homeostasis and angiogenesis, is therefore an attractive drug target for these diseases. However, there are still no drugs targeting PTP including VE-PTP. In this paper, we report the discovery of a novel VE-PTP inhibitor, Cpd-2, by fragment-based screening combining various biophysical techniques. Cpd-2 is the first VE-PTP inhibitor with a weakly acidic structure and high selectivity, unlike known strongly acidic inhibitors. We believe that this compound represents a new possibility for the development of bioavailable VE-PTP inhibitors.
Collapse
Affiliation(s)
- Wataru Asano
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Kenji Yamanaka
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yasunori Ohara
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Toru Uhara
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Satoki Doi
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Takuya Orita
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Tomoko Iwanaga
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Tsuyoshi Adachi
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Shingo Fujioka
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Tatsuo Akaki
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Kazutaka Ikegashira
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yoshiji Hantani
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| |
Collapse
|
20
|
Zhao T, He F, Zhao K, Yuxia L, Li H, Liu X, Cen J, Duan S. A Triple-Targeted Rutin-Based Self-Assembled Delivery Vector for Treating Ischemic Stroke by Vascular Normalization and Anti-Inflammation via ACE2/Ang1-7 Signaling. ACS CENTRAL SCIENCE 2023; 9:1180-1199. [PMID: 37396868 PMCID: PMC10311651 DOI: 10.1021/acscentsci.3c00377] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Indexed: 07/04/2023]
Abstract
Changes in the cerebral microenvironment caused by acute ischemic stroke-reperfusion are the main obstacle to the recovery of neurological function and an important cause of stroke recurrence after thrombolytic therapy. The intracerebral microenvironment after ischemia-reperfusion reduces the neuroplasticity of the penumbra and ultimately leads to permanent neurological damage. To overcome this challenge, we developed a triple-targeted self-assembled nanodelivery system, which combines the neuroprotective drug rutin with hyaluronic acid through esterification to form a conjugate, and then connected SS-31, a small peptide that can penetrate the blood brain barrier and target mitochondria. Brain targeting, CD44-mediated endocytosis, hyaluronidase 1-mediated degradation, and the acidic environment synergistically promoted the enrichment of nanoparticles and drug release in the injured area. Results demonstrate that rutin has a high affinity for ACE2 receptors on the cell membrane and can directly activate ACE2/Ang1-7 signaling, maintain neuroinflammation, and promote penumbra angiogenesis and normal neovascularization. Importantly, this delivery system enhanced the overall plasticity of the injured area and significantly reduced neurological damage after stroke. The relevant mechanism was expounded from the aspects of behavior, histology, and molecular cytology. All results suggest that our delivery system may be an effective and safe strategy for the treatment of acute ischemic stroke-reperfusion injury.
Collapse
Affiliation(s)
- Tingkui Zhao
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Fujin He
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Keqing Zhao
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Lin Yuxia
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Huanyu Li
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xingru Liu
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Juan Cen
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Shaofeng Duan
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan
International Joint Laboratory of Chinese Medicine Efficacy, Henan University, Kaifeng 475004, China
| |
Collapse
|
21
|
Callegari K, Dash S, Uchida H, Shingai Y, Liu C, Khodarkovskaya A, Lee Y, Ito A, Lopez A, Zhang T, Xiang J, Kluk MJ, Sanchez T. Molecular profiling of the stroke-induced alterations in the cerebral microvasculature reveals promising therapeutic candidates. Proc Natl Acad Sci U S A 2023; 120:e2205786120. [PMID: 37058487 PMCID: PMC10120001 DOI: 10.1073/pnas.2205786120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/08/2023] [Indexed: 05/11/2023] Open
Abstract
Stroke-induced cerebral microvascular dysfunction contributes to aggravation of neuronal injury and compromises the efficacy of current reperfusion therapies. Understanding the molecular alterations in cerebral microvessels in stroke will provide original opportunities for scientific investigation of novel therapeutic strategies. Toward this goal, using a recently optimized method which minimizes cell activation and preserves endothelial cell interactions and RNA integrity, we conducted a genome-wide transcriptomic analysis of cerebral microvessels in a mouse model of stroke and compared these transcriptomic alterations with the ones observed in human, nonfatal, brain stroke lesions. Results from these unbiased comparative analyses have revealed the common alterations in mouse stroke microvessels and human stroke lesions and identified shared molecular features associated with vascular disease (e.g., Serpine1/Plasminogen Activator Inhibitor-1, Hemoxygenase-1), endothelial activation (e.g., Angiopoietin-2), and alterations in sphingolipid metabolism and signaling (e.g., Sphigosine-1-Phosphate Receptor 2). Sphingolipid profiling of mouse cerebral microvessels validated the transcript data and revealed the enrichment of sphingomyelin and sphingoid species in the cerebral microvasculature compared to brain and the stroke-induced increase in ceramide species. In summary, our study has identified novel molecular alterations in several microvessel-enriched, translationally relevant, and druggable targets, which are potent modulators of endothelial function. Our comparative analyses have revealed the presence of molecular features associated with cerebral microvascular dysfunction in human chronic stroke lesions. The results shared here provide a detailed resource for therapeutic discovery of candidates for neurovascular protection in stroke and potentially, other pathologies exhibiting cerebral microvascular dysfunction.
Collapse
Affiliation(s)
- Keri Callegari
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Sabyasachi Dash
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Hiroki Uchida
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Yuto Shingai
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Catherine Liu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Anne Khodarkovskaya
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Yunkyoung Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Akira Ito
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Amanda Lopez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY10065
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY10065
| | - Michael J. Kluk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Teresa Sanchez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065
| |
Collapse
|
22
|
Miners J, van Hulle C, Ince S, Jonaitis E, Okonkwo OC, Bendlin B, Johnson S, Carlsson C, Asthana S, Love S, Blennow K, Zetterberg H. Elevated CSF angiopoietin-2 correlates with blood-brain barrier leakiness and markers of neuronal injury in early Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-2722280. [PMID: 37131622 PMCID: PMC10153378 DOI: 10.21203/rs.3.rs-2722280/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Breakdown of the neurovascular unit in early Alzheimer's disease (AD) leads to leakiness of the blood-brain barrier (BBB), contributing to cognitive decline and disease pathology. Vascular stability depends on angiopoietin-1 (ANGPT1) signalling, antagonised by angiopoietin-2 (ANGPT2) upon endothelial injury. We have examined the relationship between CSF ANGPT2 and CSF markers of BBB leakiness and disease pathology, across three independent cohorts: (i) 31 AD patients and 33 healthy controls grouped according to their biomarker profile (i.e., AD cases t-tau > 400 pg/mL, p-tau > 60 pg/mL and Aβ42 < 550 pg/mL); (ii) 121 participants in the Wisconsin Registry for Alzheimer's Prevention or Wisconsin Alzheimer's Disease Research study (84 participants cognitively unimpaired (CU) enriched for a parental history of AD, 19 participants with mild cognitive impairment (MCI), and 21 with AD); (iii) a neurologically normal cohort aged 23-78 years with paired CSF and serum samples. CSF ANGPT2 level was measured by sandwich ELISA. In cohort (i), CSF ANGPT2 was elevated in AD, correlating with CSF t-tau and p-tau181 but not Aβ42. ANGPT2 also correlated positively with CSF sPDGFRβ and fibrinogen - markers of pericyte injury and BBB leakiness. In cohort (ii), CSF ANGPT2 was highest in MCI. CSF ANGT2 correlated with CSF albumin in the CU and MCI cohorts but not in AD. ANGPT2 also correlated with t-tau and p-tau and with markers of neuronal injury (neurogranin and α-synuclein) and neuroinflammation (GFAP and YKL-40). In cohort (iii), CSF ANGPT2 correlated strongly with the CSF:serum albumin ratio. Increased CSF ANGPT2 and the CSF:serum albumin ratio showed non-significant associations with elevated serum ANGPT2 in this small cohort. Together, these data indicate that CSF ANGPT2 is associated with BBB leakiness in early AD and is closely related to tau pathology and neuronal injury. The utility of serum ANGPT2 as a biomarker of BBB damage in AD requires further study.
Collapse
Affiliation(s)
| | - Carol van Hulle
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health
| | - Selvi Ince
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health
| | - Erin Jonaitis
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health
| | - O C Okonkwo
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health
| | - Barbara Bendlin
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health
| | - Sterling Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health
| | | | | | | | | | | |
Collapse
|
23
|
McCann M, Li Y, Baccouche B, Kazlauskas A. VEGF Induces Expression of Genes That Either Promote or Limit Relaxation of the Retinal Endothelial Barrier. Int J Mol Sci 2023; 24:6402. [PMID: 37047375 PMCID: PMC10094353 DOI: 10.3390/ijms24076402] [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: 02/21/2023] [Revised: 03/21/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023] Open
Abstract
The purpose of this study was to identify genes that mediate VEGF-induced permeability. We performed RNA-Seq analysis on primary human retinal endothelial cells (HRECs) cultured in normal (5 mM) and high glucose (30 mM) conditions that were treated with vehicle, VEGF, or VEGF then anti-VEGF. We filtered our RNA-Seq dataset to identify genes with the following four characteristics: (1) regulated by VEGF, (2) VEGF regulation reversed by anti-VEGF, (3) regulated by VEGF in both normal and high glucose conditions, and (4) known contribution to vascular homeostasis. Of the resultant 18 genes, members of the Notch signaling pathway and ANGPT2 (Ang2) were selected for further study. Permeability assays revealed that while the Notch pathway was dispensable for relaxing the barrier, it contributed to maintaining an open barrier. In contrast, Ang2 limited the extent of barrier relaxation in response to VEGF. These findings indicate that VEGF engages distinct sets of genes to induce and sustain barrier relaxation. Furthermore, VEGF induces expression of genes that limit the extent of barrier relaxation. Together, these observations begin to elucidate the elegance of VEGF-mediated transcriptional regulation of permeability.
Collapse
Affiliation(s)
- Maximilian McCann
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yueru Li
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Basma Baccouche
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Andrius Kazlauskas
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
24
|
Spitzer D, Khel MI, Pütz T, Zinke J, Jia X, Sommer K, Filipski K, Thorsen F, Freiman TM, Günther S, Plate KH, Harter PN, Liebner S, Reiss Y, Di Tacchio M, Guérit S, Devraj K. A flow cytometry-based protocol for syngenic isolation of neurovascular unit cells from mouse and human tissues. Nat Protoc 2023; 18:1510-1542. [PMID: 36859615 DOI: 10.1038/s41596-023-00805-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/16/2022] [Indexed: 03/03/2023]
Abstract
The neurovascular unit (NVU), composed of endothelial cells, pericytes, juxtaposed astrocytes and microglia together with neurons, is essential for proper central nervous system functioning. The NVU critically regulates blood-brain barrier (BBB) function, which is impaired in several neurological diseases and is therefore a key therapeutic target. To understand the extent and cellular source of BBB dysfunction, simultaneous isolation and analysis of NVU cells is needed. Here, we describe a protocol for the EPAM-ia method, which is based on flow cytometry for simultaneous isolation and analysis of endothelial cells, pericytes, astrocytes and microglia. This method is based on differential processing of NVU cell types using enzymes, mechanical homogenization and filtration specific for each cell type followed by combining them for immunostaining and fluorescence-activated cell sorting. The gating strategy encompasses cell-type-specific and exclusion markers for contaminating cells to isolate the major NVU cell types. This protocol takes ~6 h for two sets of one or two animals. The isolation part requires experience in animal handling, fresh tissue processing and immunolabeling for flow cytometry. Sorted NVU cells can be used for downstream applications including transcriptomics, proteomics and cell culture. Multiple cell-type analyses using UpSet can then be applied to obtain robust targets from single or multiple NVU cell types in neurological diseases associated with BBB dysfunction. The EPAM-ia method is also amenable to isolation of several other cell types, including cancer cells and immune cells. This protocol is applicable to healthy and pathological tissue from mouse and human sources and to several cell types compared with similar protocols.
Collapse
Affiliation(s)
- Daniel Spitzer
- Department of Neurology, Goethe University, Frankfurt, Germany.,Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Maryam I Khel
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Tim Pütz
- Department of Neurology, Goethe University, Frankfurt, Germany.,Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Jenny Zinke
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Xiaoxiong Jia
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Kathleen Sommer
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Katharina Filipski
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Frits Thorsen
- The Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Thomas M Freiman
- Department of Neurosurgery, University Medical Center Rostock, Rostock, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Karl H Plate
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick N Harter
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Liebner
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany
| | - Yvonne Reiss
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Sylvaine Guérit
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Kavi Devraj
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany. .,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.
| |
Collapse
|
25
|
Shintani T, Suzuki R, Takeuchi Y, Shirasawa T, Noda M. Deletion or inhibition of PTPRO prevents ectopic fat accumulation and induces healthy obesity with markedly reduced systemic inflammation. Life Sci 2023; 313:121292. [PMID: 36535401 DOI: 10.1016/j.lfs.2022.121292] [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: 11/02/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
AIMS Chronic inflammation plays crucial roles in obesity-induced metabolic diseases. Protein tyrosine phosphatase receptor type O (PTPRO) is a member of the R3 subfamily of receptor-like protein tyrosine phosphatases. We previously suggested a role for PTPRO in the inactivation of the insulin receptor. The present study aimed to elucidate the involvement of PTPRO in the control of glucose and lipid metabolism as well as in obesity-induced systemic inflammation. MATERIALS AND METHODS Lipid accumulation in adipose tissue and the liver, the expression of inflammatory cytokines, and insulin resistance associated with systemic inflammation were investigated in hyper-obese Ptpro-KO mice by feeding a high-fat/high-sucrose diet (HFHSD). The effects of the administration of AKB9778, a specific inhibitor of PTPRO, to ob/ob mice and cultured 3T3-L1 preadipocyte cells were also examined. KEY FINDINGS Ptpro was highly expressed in visceral white adipose tissue and macrophages. Ptpro-KO mice fed HFHSD were hyper-obese, but did not have ectopic fat accumulation in the liver, dysfunctional lipid and glucose homeostasis, systemic inflammation, or insulin resistance. The administration of AKB9778 reproduced "the healthy obese phenotypes" of Ptpro-KO mice in highly obese ob/ob mice. Furthermore, the inhibition of PTPRO promoted the growth of lipid droplets in adipocytes through an increase in the phosphorylation of Tyr(117) in vimentin. SIGNIFICANCE Healthy systemic conditions with the attenuation of inflammation in hyper-obese Ptpro-KO mice were associated with the expansion of adipose tissue and low activation of NF-κb. Therefore, PTPRO may be a promising target to ameliorate hepatic steatosis and metabolic dysfunction.
Collapse
Affiliation(s)
- Takafumi Shintani
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan; Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | - Ryoko Suzuki
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | - Yasushi Takeuchi
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | | | - Masaharu Noda
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan; Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
26
|
Reiss Y, Bauer S, David B, Devraj K, Fidan E, Hattingen E, Liebner S, Melzer N, Meuth SG, Rosenow F, Rüber T, Willems LM, Plate KH. The neurovasculature as a target in temporal lobe epilepsy. Brain Pathol 2023; 33:e13147. [PMID: 36599709 PMCID: PMC10041171 DOI: 10.1111/bpa.13147] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The blood-brain barrier (BBB) is a physiological barrier maintaining a specialized brain micromilieu that is necessary for proper neuronal function. Endothelial tight junctions and specific transcellular/efflux transport systems provide a protective barrier against toxins, pathogens, and immune cells. The barrier function is critically supported by other cell types of the neurovascular unit, including pericytes, astrocytes, microglia, and interneurons. The dysfunctionality of the BBB is a hallmark of neurological diseases, such as ischemia, brain tumors, neurodegenerative diseases, infections, and autoimmune neuroinflammatory disorders. Moreover, BBB dysfunction is critically involved in epilepsy, a brain disorder characterized by spontaneously occurring seizures because of abnormally synchronized neuronal activity. While resistance to antiseizure drugs that aim to reduce neuronal hyperexcitability remains a clinical challenge, drugs targeting the neurovasculature in epilepsy patients have not been explored. The use of novel imaging techniques permits early detection of BBB leakage in epilepsy; however, the detailed mechanistic understanding of causes and consequences of BBB compromise remains unknown. Here, we discuss the current knowledge of BBB involvement in temporal lobe epilepsy with the emphasis on the neurovasculature as a therapeutic target.
Collapse
Affiliation(s)
- Yvonne Reiss
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Sebastian Bauer
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Bastian David
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Kavi Devraj
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Elif Fidan
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Elke Hattingen
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Institute of Neuroradiology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Nico Melzer
- Department of Neurology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Felix Rosenow
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Theodor Rüber
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany.,Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Laurent M Willems
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Karl H Plate
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| |
Collapse
|
27
|
Anti-osteopontin therapy leads to improved edema and infarct size in a murine model of ischemic stroke. Sci Rep 2022; 12:20925. [PMID: 36463381 PMCID: PMC9719559 DOI: 10.1038/s41598-022-25245-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/28/2022] [Indexed: 12/07/2022] Open
Abstract
Ischemic stroke is a serious neurological disorder that is associated with dysregulation of the neurovascular unit (NVU) and impairment of the blood-brain barrier (BBB). Paradoxically, reperfusion therapies can aggravate NVU and BBB dysfunction, leading to deleterious consequences in addition to the obvious benefits. Using the recently established EPAM-ia method, we identified osteopontin as a target dysregulated in multiple NVU cell types and demonstrated that osteopontin targeting in the early acute phase post-transient middle cerebral artery occlusion (tMCAO) evolves protective effects. Here, we assessed the time course of osteopontin and CD44 receptor expression in NVU cells and examined cerebroprotective effects of osteopontin targeting in early and late acute phases of ischemic stroke. Expression analysis of osteopontin and CD44 receptor post-tMCAO indicated increased levels of both, from early to late acute phases, which was supported by their co-localization in NVU cells. Combined osteopontin targeting in early and late acute phases with anti-osteopontin antibody resulted in further improvement in BBB recovery and edema reduction compared to targeting only in the early acute phase comprising the reperfusion window. Combined targeting led to reduced infarct volumes, which was not observed for the single early acute phase targeting. The effects of the therapeutic antibody were confirmed both in vitro and in vivo in reducing osteopontin and CD44 expression. Osteopontin targeting at the NVU in early and late acute phases of ischemic stroke improves edema and infarct size in mice, suggesting anti-osteopontin therapy as promising adjunctive treatment to reperfusion therapy.
Collapse
|
28
|
Collazos-Alemán JD, Gnecco-González S, Jaramillo-Zarama B, Jiménez-Mora MA, Mendivil CO. The Role of Angiopoietins in Neovascular Diabetes-Related Retinal Diseases. Diabetes Ther 2022; 13:1811-1821. [PMID: 36331711 PMCID: PMC9663771 DOI: 10.1007/s13300-022-01326-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Diabetic retinopathy is a devastating and frequent complication of poorly controlled diabetes, whose pathogenesis is still only partially understood. Advances in basic research over the last two decades have led to the discovery of angiopoietins, proteins that strongly influence the growth and integrity of blood vessels in many vascular beds, with particular importance in the retina. Angiopoietin 1 (Ang1), produced mostly by pericytes and platelets, and angiopoietin 2 (Ang2), produced mainly by endothelial cells, bind to the same receptor (Tie2), but exert opposing effects on target cells. Ang1 maintains the stability of the mature vasculature, while Ang2 promotes vessel wall destabilization and disruption of the connections between endothelial cells and pericytes. Human retinal endothelial cells exposed to Ang2 show reduced membrane expression of the adhesion molecule VE-cadherin, and patients with proliferative diabetic retinopathy or diabetic macular edema have markedly increased vitreal concentrations of Ang2. Faricimab, a bi-specific antibody simultaneously directed against Ang2 and VEGF, has shown promising results in clinical trials among patients with diabetic retinopathy, and other agents targeting the angiopoietin system are currently in development.
Collapse
Affiliation(s)
| | - Sofía Gnecco-González
- School of Medicine, Universidad de los Andes, Carrera 7 No 116-05, Of 413, Bogotá, Colombia
| | | | - Mario A Jiménez-Mora
- Department of Ophthalmology, Faculty of Medicine, National University of Colombia, Bogotá, Colombia
| | - Carlos O Mendivil
- School of Medicine, Universidad de los Andes, Carrera 7 No 116-05, Of 413, Bogotá, Colombia.
- Section of Endocrinology, Fundación Santa Fe de Bogotá, Bogotá, Colombia.
| |
Collapse
|
29
|
Moxon JV, Kraeuter AK, Phie J, Juliano S, Anderson G, Standley G, Sealey C, White RP, Golledge J. Serum angiopoietin-1 concentration does not distinguish patients with ischaemic stroke from those presenting to hospital with ischaemic stroke mimics. BMC Cardiovasc Disord 2022; 22:462. [PMID: 36333663 PMCID: PMC9636674 DOI: 10.1186/s12872-022-02918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Background A previous study found that circulating angiopoietin-1 (angpt-1) concentrations were significantly lower in patients who had a recent ischaemic stroke compared to healthy controls. The primary aim of this study was to assess whether serum angpt-1 could be used as a diagnostic test of ischemic stroke in patients presenting to hospital as an emergency. Exploratory analyses investigated the association of proteins functionally related to angpt-1 (angpt-2, Tie-2, matrix metalloproteinase-9 and vascular endothelial growth factors A, C and D) with ischaemic stroke diagnosis. Methods Patients presenting to Townsville University Hospital for emergency assessment of stroke-like symptoms were consecutively recruited and provided a blood sample. After assessment by a consultant neurologist, patients were grouped into those who did, or did not have ischaemic stroke. The potential for serum angpt-1 to diagnose ischaemic stroke was assessed using receiver operator characteristic (ROC) curves. Cross-sectional analyses appraised inter-group differences in the serum concentration of other proteins. Results One-hundred and twenty-six patients presenting to Townsville University Hospital for emergency assessment of stroke-like symptoms were recruited (median time from symptom onset to hospital presentation: 2.6 (inter-quartile range: 1.2–4.6) hours). Serum angpt-1 had poor ability to diagnose ischaemic stroke in analyses using the whole cohort, or in sensitivity analyses (area under the ROC curve 0.51 (95% CI: 0.41–0.62) and 0.52 (95% CI: 0.39–0.64), respectively). No associations of serum angpt-1 concentration with ischaemic stroke severity, symptom duration or aetiology were observed. Serum concentrations of the other assessed proteins did not differ between patient groups. Conclusions Serum angpt-1 concentration is unlikely to be useful for emergency diagnosis of ischaemic stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02918-w.
Collapse
|
30
|
Goncalves A, Antonetti DA. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids Barriers CNS 2022; 19:86. [PMID: 36320068 PMCID: PMC9628113 DOI: 10.1186/s12987-022-00386-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
The unique environment of the brain and retina is tightly regulated by blood-brain barrier and the blood-retinal barrier, respectively, to ensure proper neuronal function. Endothelial cells within these tissues possess distinct properties that allow for controlled passage of solutes and fluids. Pericytes, glia cells and neurons signal to endothelial cells (ECs) to form and maintain the barriers and control blood flow, helping to create the neurovascular unit. This barrier is lost in a wide range of diseases affecting the central nervous system (CNS) and retina such as brain tumors, stroke, dementia, and in the eye, diabetic retinopathy, retinal vein occlusions and age-related macular degeneration to name prominent examples. Recent studies directly link barrier changes to promotion of disease pathology and degradation of neuronal function. Understanding how these barriers form and how to restore these barriers in disease provides an important point for therapeutic intervention. This review aims to describe the fundamentals of the blood-tissue barriers of the CNS and how the use of transgenic animal models led to our current understanding of the molecular framework of these barriers. The review also highlights examples of targeting barrier properties to protect neuronal function in disease states.
Collapse
Affiliation(s)
- Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA.
| |
Collapse
|
31
|
Lv LL, Du YT, Chen X, Lei Y, Sun FY. Neuroprotective Effect of Angiopoietin2 Is Associated with Angiogenesis in Mouse Brain Following Ischemic Stroke. Brain Sci 2022; 12:1428. [PMID: 36358355 PMCID: PMC9688484 DOI: 10.3390/brainsci12111428] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 07/30/2023] Open
Abstract
Angiogenic factors play an important role in protecting, repairing, and reconstructing vessels after ischemic stroke. In the brains of transient focal cerebral ischemic mice, we observed a reduction in infarct volume after the administration of Angiopoietin 2 (Angpt2), but whether this process is promoted by Angpt2-induced angiogenesis has not been fully elaborated. Therefore, this study explored the angiogenic activities, in reference to CD34 which is a marker of activated ECs and blood vessels, of cultured ECs in vitro and in ischemic damaged cerebral area in mice following Angpt2 administration. Our results demonstrate that Angpt2 administration (100 ng/mL) is neuroprotective by significantly increasing the CD34 expression in in vitro-cultured ECs, reducing the infarct volume and mitigating neuronal loss, as well as enhancing CD34+ vascular length and area. In conclusion, these results indicate that Angpt2 promotes repair and attenuates ischemic injury, and that the mechanism of this is closely associated with angiogenesis in the brain after stroke.
Collapse
Affiliation(s)
- Ling-Ling Lv
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Hanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Ting Du
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Hanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiao Chen
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Hanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Lei
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Hanghai Medical College, Fudan University, Shanghai 200032, China
| | - Feng-Yan Sun
- Department of Neurobiology and State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institute for Basic Research on Aging and Medicine of School of Basic Medical Sciences and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Hanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
32
|
Yang J, Ran M, Li H, Lin Y, Ma K, Yang Y, Fu X, Yang S. New insight into neurological degeneration: Inflammatory cytokines and blood–brain barrier. Front Mol Neurosci 2022; 15:1013933. [PMID: 36353359 PMCID: PMC9637688 DOI: 10.3389/fnmol.2022.1013933] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Neurological degeneration after neuroinflammation, such as that resulting from Alzheimer’s disease (AD), stroke, multiple sclerosis (MS), and post-traumatic brain injury (TBI), is typically associated with high mortality and morbidity and with permanent cognitive dysfunction, which places a heavy economic burden on families and society. Diagnosing and curing these diseases in their early stages remains a challenge for clinical investigation and treatment. Recent insight into the onset and progression of these diseases highlights the permeability of the blood–brain barrier (BBB). The primary factor that influences BBB structure and function is inflammation, especially the main cytokines including IL-1β, TNFα, and IL-6, the mechanism on the disruption of which are critical component of the aforementioned diseases. Surprisingly, the main cytokines from systematic inflammation can also induce as much worse as from neurological diseases or injuries do. In this review, we will therefore discuss the physiological structure of BBB, the main cytokines including IL-1β, TNFα, IL-6, and their mechanism on the disruption of BBB and recent research about the main cytokines from systematic inflammation inducing the disruption of BBB and cognitive impairment, and we will eventually discuss the need to prevent the disruption of BBB.
Collapse
Affiliation(s)
- Jie Yang
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Mingzi Ran
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Anaesthesiology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Hongyu Li
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Ye Lin
- Department of Neurology, The First Medical Centre, PLA General Hospital, Beijing, China
| | - Kui Ma
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
| | - Yuguang Yang
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Xiaobing Fu,
| | - Siming Yang
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
- *Correspondence: Siming Yang,
| |
Collapse
|
33
|
Klepper S, Jung S, Dittmann L, Geppert CI, Hartmann A, Beier N, Trollmann R. Further Evidence of Neuroprotective Effects of Recombinant Human Erythropoietin and Growth Hormone in Hypoxic Brain Injury in Neonatal Mice. Int J Mol Sci 2022; 23:ijms23158693. [PMID: 35955834 PMCID: PMC9368903 DOI: 10.3390/ijms23158693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Experimental in vivo data have recently shown complementary neuroprotective actions of rhEPO and growth hormone (rhGH) in a neonatal murine model of hypoxic brain injury. Here, we hypothesized that rhGH and rhEPO mediate stabilization of the blood−brain barrier (BBB) and regenerative vascular effects in hypoxic injury to the developing brain. Using an established model of neonatal hypoxia, neonatal mice (P7) were treated i.p. with rhGH (4000 µg/kg) or rhEPO (5000 IU/kg) 0/12/24 h after hypoxic exposure. After a regeneration period of 48 h or 7 d, cerebral mRNA expression of Vegf-A, its receptors and co-receptors, and selected tight junction proteins were determined using qRT-PCR and ELISA. Vessel structures were assessed by Pecam-1 and occludin (Ocln) IHC. While Vegf-A expression increased significantly with rhGH treatment (p < 0.01), expression of the Vegfr and TEK receptor tyrosine kinase (Tie-2) system remained unchanged. RhEPO increased Vegf-A (p < 0.05) and Angpt-2 (p < 0.05) expression. While hypoxia reduced the mean vessel area in the parietal cortex compared to controls (p < 0.05), rhGH and rhEPO prevented this reduction after 48 h of regeneration. Hypoxia significantly reduced the Ocln+ fraction of cortical vascular endothelial cells. Ocln signal intensity increased in the cortex in response to rhGH (p < 0.05) and in the cortex and hippocampus in response to rhEPO (p < 0.05). Our data indicate that rhGH and rhEPO have protective effects on hypoxia-induced BBB disruption and regenerative vascular effects during the post-hypoxic period in the developing brain.
Collapse
Affiliation(s)
- Simon Klepper
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Susan Jung
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Lara Dittmann
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Carol I. Geppert
- Institute of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg, Krankenhausstr. 8, 91054 Erlangen, Germany
| | - Arnd Hartmann
- Institute of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg, Krankenhausstr. 8, 91054 Erlangen, Germany
| | - Nicole Beier
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
| | - Regina Trollmann
- Division of Pediatric Neurology, Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestr. 15, 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8533753; Fax: +49-9131-8533389
| |
Collapse
|
34
|
Smith-Cohn MA, Burley NB, Grossman SA. Transient Opening of the Blood-Brain Barrier by Vasoactive Peptides to Increase CNS Drug Delivery: Reality Versus Wishful Thinking? Curr Neuropharmacol 2022; 20:1383-1399. [PMID: 35100958 PMCID: PMC9881081 DOI: 10.2174/1570159x20999220131163504] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/02/2021] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The blood-brain barrier inhibits the central nervous system penetration of 98% of small molecule drugs and virtually all biologic agents, which has limited progress in treating neurologic disease. Vasoactive peptides have been shown in animal studies to transiently disrupt the blood-brain barrier and regadenoson is currently being studied in humans to determine if it can improve drug delivery to the brain. However, many other vasoactive peptides could potentially be used for this purpose. METHODS We performed a review of the literature evaluating the physiologic effects of vasoactive peptides on the vasculature of the brain and systemic organs. To assess the likelihood that a vasoactive peptide might transiently disrupt the blood-brain barrier, we devised a four-tier classification system to organize the available evidence. RESULTS We identified 32 vasoactive peptides with potential blood-brain barrier permeabilityaltering properties. To date, none of these are shown to open the blood-brain barrier in humans. Twelve vasoactive peptides increased blood-brain barrier permeability in rodents. The remaining 20 had favorable physiologic effects on blood vessels but lacked specific information on permeability changes to the blood-brain barrier. CONCLUSION Vasoactive peptides remain an understudied class of drugs with the potential to increase drug delivery and improve treatment in patients with brain tumors and other neurologic diseases. Dozens of vasoactive peptides have yet to be formally evaluated for this important clinical effect. This narrative review summarizes the available data on vasoactive peptides, highlighting agents that deserve further in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Matthew A. Smith-Cohn
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA; ,Address correspondence to these authors at the The Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Swedish Health Services, 500 17th Ave, James Tower, Suite 540, Seattle, WA 98122, USA; Tel: 206-320-2300; Fax: 206-320-8149; E-mail: , Sidney Kimmel Cancer Center, Skip Viragh Building, 201 North Broadway, 9th Floor (Mailbox #3), Baltimore, MD 21287, USA; E-mail:
| | - Nicholas B. Burley
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA;
| | - Stuart A. Grossman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA,Address correspondence to these authors at the The Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Swedish Health Services, 500 17th Ave, James Tower, Suite 540, Seattle, WA 98122, USA; Tel: 206-320-2300; Fax: 206-320-8149; E-mail: , Sidney Kimmel Cancer Center, Skip Viragh Building, 201 North Broadway, 9th Floor (Mailbox #3), Baltimore, MD 21287, USA; E-mail:
| |
Collapse
|
35
|
Wang R, Yang M, Jiang L, Huang M. Role of Angiopoietin-Tie axis in vascular and lymphatic systems and therapeutic interventions. Pharmacol Res 2022; 182:106331. [PMID: 35772646 DOI: 10.1016/j.phrs.2022.106331] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/11/2022] [Accepted: 06/24/2022] [Indexed: 12/29/2022]
Abstract
The Angiopoietin (Ang)-Tyrosine kinase with immunoglobulin-like and EGF-like domains (Tie) axis is an endothelial cell-specific ligand-receptor signaling pathway necessary for vascular and lymphatic development. The Ang-Tie axis is involved in regulating angiogenesis, vascular remodeling, vascular permeability, and inflammation to maintain vascular quiescence. Disruptions in the Ang-Tie axis are involved in many vascular and lymphatic system diseases and play an important role in physiological and pathological vascular conditions. Given recent advances in the Ang-Tie axis in the vascular and lymphatic systems, this review focuses on the multiple functions of the Ang-Tie axis in inflammation-induced vascular permeability, vascular remodeling, atherosclerosis, ocular angiogenesis, tumor angiogenesis, and metastasis. A summary of relevant therapeutic approaches to the Ang-Tie axis, including therapeutic antibodies, recombinant proteins and small molecule drugs are also discussed. The purpose of this review is to provide new hypotheses and identify potential therapeutic strategies based on the Ang-Tie signaling axis for the treatment of vascular and lymphatic-related diseases.
Collapse
Affiliation(s)
- Rui Wang
- College of Chemistry, Fuzhou University, Fuzhou 350116, Fujian, China
| | - Moua Yang
- Division of Hemostasis & Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA02215, United States
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, Fujian, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, Fujian, China.
| |
Collapse
|
36
|
Nehra G, Bauer B, Hartz AMS. Blood-brain barrier leakage in Alzheimer's disease: From discovery to clinical relevance. Pharmacol Ther 2022; 234:108119. [PMID: 35108575 PMCID: PMC9107516 DOI: 10.1016/j.pharmthera.2022.108119] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD brain pathology starts decades before the onset of clinical symptoms. One early pathological hallmark is blood-brain barrier dysfunction characterized by barrier leakage and associated with cognitive decline. In this review, we summarize the existing literature on the extent and clinical relevance of barrier leakage in AD. First, we focus on AD animal models and their susceptibility to barrier leakage based on age and genetic background. Second, we re-examine barrier dysfunction in clinical and postmortem studies, summarize changes that lead to barrier leakage in patients and highlight the clinical relevance of barrier leakage in AD. Third, we summarize signaling mechanisms that link barrier leakage to neurodegeneration and cognitive decline in AD. Finally, we discuss clinical relevance and potential therapeutic strategies and provide future perspectives on investigating barrier leakage in AD. Identifying mechanistic steps underlying barrier leakage has the potential to unravel new targets that can be used to develop novel therapeutic strategies to repair barrier leakage and slow cognitive decline in AD and AD-related dementias.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
37
|
Kam JY, Cheng T, Garland DC, Britton WJ, Tobin DM, Oehlers SH. Inhibition of infection-induced vascular permeability modulates host leukocyte recruitment to Mycobacterium marinum granulomas in zebrafish. Pathog Dis 2022; 80:ftac009. [PMID: 35438161 PMCID: PMC9053305 DOI: 10.1093/femspd/ftac009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/23/2022] [Accepted: 04/14/2022] [Indexed: 01/07/2023] Open
Abstract
Mycobacterial granuloma formation involves significant stromal remodeling including the growth of leaky, granuloma-associated vasculature. These permeable blood vessels aid mycobacterial growth, as antiangiogenic or vascular normalizing therapies are beneficial host-directed therapies in preclinical models of tuberculosis across host-mycobacterial pairings. Using the zebrafish-Mycobacterium marinum infection model, we demonstrate that vascular normalization by inhibition of vascular endothelial protein tyrosine phosphatase (VE-PTP) decreases granuloma hypoxia, the opposite effect of hypoxia-inducing antiangiogenic therapy. Inhibition of VE-PTP decreased neutrophil recruitment to granulomas in adult and larval zebrafish, and decreased the proportion of neutrophils that extravasated distal to granulomas. Furthermore, VE-PTP inhibition increased the accumulation of T cells at M. marinum granulomas. Our study provides evidence that, similar to the effect in solid tumors, vascular normalization during mycobacterial infection increases the T cell:neutrophil ratio in lesions which may be correlates of protective immunity.
Collapse
Affiliation(s)
- Julia Y Kam
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Tina Cheng
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Danielle C Garland
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
- The University of Sydney, Sydney Institute for Infectious Diseases, Camperdown, NSW 2050, Australia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science Technology and Research (A*STAR), Singapore, 138648, Singapore
| |
Collapse
|
38
|
Zhou SY, Guo ZN, Zhang DH, Qu Y, Jin H. The Role of Pericytes in Ischemic Stroke: Fom Cellular Functions to Therapeutic Targets. Front Mol Neurosci 2022; 15:866700. [PMID: 35493333 PMCID: PMC9043812 DOI: 10.3389/fnmol.2022.866700] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
Ischemic stroke (IS) is a cerebrovascular disease causing high rates of disability and fatality. In recent years, the concept of the neurovascular unit (NVU) has been accepted by an increasing number of researchers and is expected to become a new paradigm for exploring the pathogenesis and treatment of IS. NVUs are composed of neurons, endothelial cells, pericytes, astrocytes, microglia, and the extracellular matrix. As an important part of the NVU, pericytes provide support for other cellular components and perform a variety of functions, including participating in the maintenance of the normal physiological function of the blood–brain barrier, regulating blood flow, and playing a role in inflammation, angiogenesis, and neurogenesis. Therefore, treatment strategies targeting pericyte functions, regulating pericyte epigenetics, and transplanting pericytes warrant exploration. In this review, we describe the reactions of pericytes after IS, summarize the potential therapeutic targets and strategies targeting pericytes for IS, and provide new treatment ideas for ischemic stroke.
Collapse
Affiliation(s)
- Sheng-Yu Zhou
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Zhen-Ni Guo
- Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Dian-Hui Zhang
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Qu
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Hang Jin,
| |
Collapse
|
39
|
Xiong M, Feng Y, Huang S, Lv S, Deng Y, Li M, Wang P, Luo M, Wen H, Zhang W. Teriparatide induces angiogenesis in ischemic cerebral infarction zones of rats through AC/PKA signaling and reduces ischemia-reperfusion injury. Biomed Pharmacother 2022; 148:112728. [PMID: 35220030 DOI: 10.1016/j.biopha.2022.112728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/02/2022] Open
Abstract
Teriparatide is a commonly used drug indicated for the treatment of osteoporosis in postmenopausal women. Teriparatide can also upregulate Ang-1 expression through the AC/PKA signaling pathway to promote angiogenesis. At present, promoting angiogenesis is a promising but unrealized strategy for the treatment of ischemic cerebral infarction. However, there are few studies on the application of teriparatide in the treatment of cerebral infarction. We used teriparatide to treat ischemic cerebral infarction in rats and obtained three major findings. First, teriparatide can promote angiogenesis, reduce cerebral infarct size, and increase cerebral perfusion by upregulating Ang-1 expression. Second, teriparatide can promote the expression of HO1, SOD2 and inhibit the production of pro-inflammatory cytokines IL-1β, IL-6 by upregulating Nrf2 expression. Third, we further found that teriparatide can mitigate blood-brain barrier disruption and brain edema by downregulating the expressions of MMP9, Ang-2 and AQP4. Our results indicate that teriparatide is neuroprotective through multiple mechanisms of action that include promoting angiogenesis, inhibiting oxidative stress and neuroinflammation, protecting blood-brain barrier, and reducing brain edema.
Collapse
Affiliation(s)
- Moliang Xiong
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Yun Feng
- Department of Pediatrics, Hospital of the 74th Group Army of the Chinese people's Liberation Army, Guangzhou 510282, China
| | - Shujie Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Siyuan Lv
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Yuhao Deng
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Min Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Pengfei Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Minjie Luo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Huangtao Wen
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China
| | - Wangming Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510282, China.
| |
Collapse
|
40
|
Identification of Novel Circulating miRNAs in Patients with Acute Ischemic Stroke. Int J Mol Sci 2022; 23:ijms23063387. [PMID: 35328807 PMCID: PMC8955546 DOI: 10.3390/ijms23063387] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic strokes are associated with significant morbidity and mortality, but currently there are no reliable prognostic or diagnostic blood biomarkers. MicroRNAs (miRNAs) regulate various molecular pathways and may be used as biomarkers. Using RNA-Seq, we conducted comprehensive circulating miRNA profiling in patients with ischemic stroke compared with healthy controls. Samples were collected within 24 h of clinical diagnosis. Stringent analysis criteria of discovery (46 cases and 95 controls) and validation (47 cases and 96 controls) cohorts led to the identification of 10 differentially regulated miRNAs, including 5 novel miRNAs, with potential diagnostic significance. Hsa-miR-451a was the most significantly upregulated miRNA (FC; 4.8, FDR; 3.78 × 10−85), while downregulated miRNAs included hsa-miR-574-5p and hsa-miR-142-3p, among others. Importantly, we computed a multivariate classifier based on the identified miRNA panel to differentiate between ischemic stroke patients and healthy controls, which showed remarkably high sensitivity (0.94) and specificity (0.99). The area under the ROC curve was 0.97 and it is superior to other current available biomarkers. Moreover, in samples collected one month following stroke, we found sustained upregulation of hsa-miR-451a and downregulation of another 5 miRNAs. Lastly, we report 3 miRNAs that were significantly associated with poor clinical outcomes of stroke, as defined by the modified Rankin scores. The clinical translation of the identified miRNA panel may be explored further.
Collapse
|
41
|
Martin M, Vermeiren S, Bostaille N, Eubelen M, Spitzer D, Vermeersch M, Profaci CP, Pozuelo E, Toussay X, Raman-Nair J, Tebabi P, America M, De Groote A, Sanderson LE, Cabochette P, Germano RFV, Torres D, Boutry S, de Kerchove d'Exaerde A, Bellefroid EJ, Phoenix TN, Devraj K, Lacoste B, Daneman R, Liebner S, Vanhollebeke B. Engineered Wnt ligands enable blood-brain barrier repair in neurological disorders. Science 2022; 375:eabm4459. [PMID: 35175798 DOI: 10.1126/science.abm4459] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The blood-brain barrier (BBB) protects the central nervous system (CNS) from harmful blood-borne factors. Although BBB dysfunction is a hallmark of several neurological disorders, therapies to restore BBB function are lacking. An attractive strategy is to repurpose developmental BBB regulators, such as Wnt7a, into BBB-protective agents. However, safe therapeutic use of Wnt ligands is complicated by their pleiotropic Frizzled signaling activities. Taking advantage of the Wnt7a/b-specific Gpr124/Reck co-receptor complex, we genetically engineered Wnt7a ligands into BBB-specific Wnt activators. In a "hit-and-run" adeno-associated virus-assisted CNS gene delivery setting, these new Gpr124/Reck-specific agonists protected BBB function, thereby mitigating glioblastoma expansion and ischemic stroke infarction. This work reveals that the signaling specificity of Wnt ligands is adjustable and defines a modality to treat CNS disorders by normalizing the BBB.
Collapse
Affiliation(s)
- Maud Martin
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Simon Vermeiren
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Naguissa Bostaille
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Marie Eubelen
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Daniel Spitzer
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Université de Mons, Gosselies B-6041, Belgium
| | - Caterina P Profaci
- Departments of Pharmacology and Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Elisa Pozuelo
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université libre de Bruxelles, Brussels B-1070, Belgium
| | - Xavier Toussay
- Ottawa Hospital Research Institute, Neuroscience Program, Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Faculty of Medicine, Ottawa, Ontario, Canada
| | - Joanna Raman-Nair
- Ottawa Hospital Research Institute, Neuroscience Program, Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Faculty of Medicine, Ottawa, Ontario, Canada
| | - Patricia Tebabi
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Michelle America
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Aurélie De Groote
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université libre de Bruxelles, Brussels B-1070, Belgium
| | - Leslie E Sanderson
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Pauline Cabochette
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Raoul F V Germano
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - David Torres
- Institut d'Immunologie Médicale, Université libre de Bruxelles, Gosselies, Belgium
| | - Sébastien Boutry
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Université de Mons, Gosselies B-6041, Belgium
| | - Alban de Kerchove d'Exaerde
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université libre de Bruxelles, Brussels B-1070, Belgium
| | - Eric J Bellefroid
- Laboratory of Developmental Genetics, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Kavi Devraj
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Faculty of Medicine, Ottawa, Ontario, Canada
| | - Richard Daneman
- Departments of Pharmacology and Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies B-6041, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
| |
Collapse
|
42
|
Kellar D, Register T, Lockhart SN, Aisen P, Raman R, Rissman RA, Brewer J, Craft S. Intranasal insulin modulates cerebrospinal fluid markers of neuroinflammation in mild cognitive impairment and Alzheimer's disease: a randomized trial. Sci Rep 2022; 12:1346. [PMID: 35079029 PMCID: PMC8789895 DOI: 10.1038/s41598-022-05165-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Intranasal insulin (INI) has shown promise as a treatment for Alzheimer's disease (AD) in pilot clinical trials. In a recent phase 2 trial, participants with mild cognitive impairment (MCI) or AD who were treated with INI with one of two delivery devices showed improved cerebral spinal fluid (CSF) biomarker profiles and slower symptom progression compared with placebo. In the cohort which showed benefit, we measured changes in CSF markers of inflammation, immune function and vascular integrity and assessed their relationship with changes in cognition, brain volume, and CSF amyloid and tau concentrations. The insulin-treated group had increased CSF interferon-γ (p = 0.032) and eotaxin (p = 0.049), and reduced interleukin-6 (p = 0.048) over the 12 month trial compared to placebo. Trends were observed for increased CSF macrophage-derived chemokine for the placebo group (p = 0.083), and increased interleukin-2 in the insulin-treated group (p = 0.093). Insulin-treated and placebo groups showed strikingly different patterns of associations between changes in CSF immune/inflammatory/vascular markers and changes in cognition, brain volume, and amyloid and tau concentrations. In summary, INI treatment altered the typical progression of markers of inflammation and immune function seen in AD, suggesting that INI may promote a compensatory immune response associated with therapeutic benefit.
Collapse
Affiliation(s)
- Derek Kellar
- Department of Internal Medicine-Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Thomas Register
- Department of Internal Medicine-Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Samuel N Lockhart
- Department of Internal Medicine-Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Paul Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, USA
| | - Rema Raman
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, USA
| | - Robert A Rissman
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, USA
| | - James Brewer
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, USA
| | - Suzanne Craft
- Department of Internal Medicine-Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
43
|
Spitzer D, Guérit S, Puetz T, Khel MI, Armbrust M, Dunst M, Macas J, Zinke J, Devraj G, Jia X, Croll F, Sommer K, Filipski K, Freiman TM, Looso M, Günther S, Di Tacchio M, Plate KH, Reiss Y, Liebner S, Harter PN, Devraj K. Profiling the neurovascular unit unveils detrimental effects of osteopontin on the blood-brain barrier in acute ischemic stroke. Acta Neuropathol 2022; 144:305-337. [PMID: 35752654 PMCID: PMC9288377 DOI: 10.1007/s00401-022-02452-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/01/2022]
Abstract
Blood-brain barrier (BBB) dysfunction, characterized by degradation of BBB junctional proteins and increased permeability, is a crucial pathophysiological feature of acute ischemic stroke. Dysregulation of multiple neurovascular unit (NVU) cell types is involved in BBB breakdown in ischemic stroke that may be further aggravated by reperfusion therapy. Therefore, therapeutic co-targeting of dysregulated NVU cell types in acute ischemic stroke constitutes a promising strategy to preserve BBB function and improve clinical outcome. However, methods for simultaneous isolation of multiple NVU cell types from the same diseased central nervous system (CNS) tissue, crucial for the identification of therapeutic targets in dysregulated NVU cells, are lacking. Here, we present the EPAM-ia method, that facilitates simultaneous isolation and analysis of the major NVU cell types (endothelial cells, pericytes, astrocytes and microglia) for the identification of therapeutic targets in dysregulated NVU cells to improve the BBB function. Applying this method, we obtained a high yield of pure NVU cells from murine ischemic brain tissue, and generated a valuable NVU transcriptome database ( https://bioinformatics.mpi-bn.mpg.de/SGD_Stroke ). Dissection of the NVU transcriptome revealed Spp1, encoding for osteopontin, to be highly upregulated in all NVU cells 24 h after ischemic stroke. Upregulation of osteopontin was confirmed in stroke patients by immunostaining, which was comparable with that in mice. Therapeutic targeting by subcutaneous injection of an anti-osteopontin antibody post-ischemic stroke in mice resulted in neutralization of osteopontin expression in the NVU cell types investigated. Apart from attenuated glial activation, osteopontin neutralization was associated with BBB preservation along with decreased brain edema and reduced risk for hemorrhagic transformation, resulting in improved neurological outcome and survival. This was supported by BBB-impairing effects of osteopontin in vitro. The clinical significance of these findings is that anti-osteopontin antibody therapy might augment current approved reperfusion therapies in acute ischemic stroke by minimizing deleterious effects of ischemia-induced BBB disruption.
Collapse
Affiliation(s)
- Daniel Spitzer
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,Department of Neurology, University Hospital, Goethe University, 60528 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528 Frankfurt, Germany
| | - Sylvaine Guérit
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Tim Puetz
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,Department of Neurology, University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Maryam I. Khel
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Moritz Armbrust
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Maika Dunst
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Jadranka Macas
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Jenny Zinke
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Gayatri Devraj
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Xiaoxiong Jia
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Florian Croll
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Kathleen Sommer
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Katharina Filipski
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany ,grid.511198.5Frankfurt Cancer Institute (FCI), 60528 Frankfurt, Germany
| | - Thomas M. Freiman
- grid.413108.f0000 0000 9737 0454Department of Neurosurgery, University Medical Center Rostock, 18057 Rostock, Germany ,grid.7839.50000 0004 1936 9721LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528 Frankfurt, Germany
| | - Mario Looso
- grid.418032.c0000 0004 0491 220XMax Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Stefan Günther
- grid.418032.c0000 0004 0491 220XMax Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Mariangela Di Tacchio
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany
| | - Karl-Heinz Plate
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany ,grid.511198.5Frankfurt Cancer Institute (FCI), 60528 Frankfurt, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, 60528 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528 Frankfurt, Germany
| | - Yvonne Reiss
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany ,grid.511198.5Frankfurt Cancer Institute (FCI), 60528 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528 Frankfurt, Germany
| | - Stefan Liebner
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, 60528 Frankfurt, Germany ,Excellence Cluster Cardio Pulmonary System (CPI), Partner Site Frankfurt, 60528 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528 Frankfurt, Germany
| | - Patrick N. Harter
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, 60528 Frankfurt, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany ,grid.511198.5Frankfurt Cancer Institute (FCI), 60528 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528 Frankfurt, Germany
| | - Kavi Devraj
- Edinger Institute (Institute of Neurology), University Hospital, Goethe University, 60528, Frankfurt, Germany. .,Frankfurt Cancer Institute (FCI), 60528, Frankfurt, Germany. .,LOEWE - Center for Personalized Translational Epilepsy Research (CePTER), Goethe University, 60528, Frankfurt, Germany.
| |
Collapse
|
44
|
Amelio GS, Provitera L, Raffaeli G, Tripodi M, Amodeo I, Gulden S, Cortesi V, Manzoni F, Cervellini G, Tomaselli A, Pravatà V, Garrido F, Villamor E, Mosca F, Cavallaro G. Endothelial dysfunction in preterm infants: The hidden legacy of uteroplacental pathologies. Front Pediatr 2022; 10:1041919. [PMID: 36405831 PMCID: PMC9671930 DOI: 10.3389/fped.2022.1041919] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Millions of infants are born prematurely every year worldwide. Prematurity, particularly at lower gestational ages, is associated with high mortality and morbidity and is a significant global health burden. Pregnancy complications and preterm birth syndrome strongly impact neonatal clinical phenotypes and outcomes. The vascular endothelium is a pivotal regulator of fetal growth and development. In recent years, the key role of uteroplacental pathologies impairing endothelial homeostasis is emerging. Conditions leading to very and extremely preterm birth can be classified into two main pathophysiological patterns or endotypes: infection/inflammation and dysfunctional placentation. The first is frequently related to chorioamnionitis, whereas the second is commonly associated with hypertensive disorders of pregnancy and fetal growth restriction. The nature, timing, and extent of prenatal noxa may alter fetal and neonatal endothelial phenotype and functions. Changes in the luminal surface, oxidative stress, growth factors imbalance, and dysregulation of permeability and vascular tone are the leading causes of endothelial dysfunction in preterm infants. However, the available evidence regarding endothelial physiology and damage is limited in neonates compared to adults. Herein, we discuss the current knowledge on endothelial dysfunction in the infectious/inflammatory and dysfunctional placentation endotypes of prematurity, summarizing their molecular features, available biomarkers, and clinical impact. Furthermore, knowledge gaps, shadows, and future research perspectives are highlighted.
Collapse
Affiliation(s)
- Giacomo Simeone Amelio
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Gulden
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Cortesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Francesca Manzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Gaia Cervellini
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Valentina Pravatà
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, Madrid, Spain
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), University of Maastricht, Maastricht, Netherlands
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
45
|
Modulation of the Blood-Brain Barrier for Drug Delivery to Brain. Pharmaceutics 2021; 13:pharmaceutics13122024. [PMID: 34959306 PMCID: PMC8708282 DOI: 10.3390/pharmaceutics13122024] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
The blood-brain barrier (BBB) precisely controls brain microenvironment and neural activity by regulating substance transport into and out of the brain. However, it severely hinders drug entry into the brain, and the efficiency of various systemic therapies against brain diseases. Modulation of the BBB via opening tight junctions, inhibiting active efflux and/or enhancing transcytosis, possesses the potential to increase BBB permeability and improve intracranial drug concentrations and systemic therapeutic efficiency. Various strategies of BBB modulation have been reported and investigated preclinically and/or clinically. This review describes conventional and emerging BBB modulation strategies and related mechanisms, and safety issues according to BBB structures and functions, to try to give more promising directions for designing more reasonable preclinical and clinical studies.
Collapse
|
46
|
Li X, Vemireddy V, Cai Q, Xiong H, Kang P, Li X, Giannotta M, Hayenga HN, Pan E, Sirsi SR, Mateo C, Kleinfeld D, Greene C, Campbell M, Dejana E, Bachoo R, Qin Z. Reversibly Modulating the Blood-Brain Barrier by Laser Stimulation of Molecular-Targeted Nanoparticles. NANO LETTERS 2021; 21:9805-9815. [PMID: 34516144 PMCID: PMC8616836 DOI: 10.1021/acs.nanolett.1c02996] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The blood-brain barrier (BBB) is highly selective and acts as the interface between the central nervous system and circulation. While the BBB is critical for maintaining brain homeostasis, it represents a formidable challenge for drug delivery. Here we synthesized gold nanoparticles (AuNPs) for targeting the tight junction specifically and demonstrated that transcranial picosecond laser stimulation of these AuNPs post intravenous injection increases the BBB permeability. The BBB permeability change can be graded by laser intensity, is entirely reversible, and involves increased paracellular diffusion. BBB modulation does not lead to significant disruption in the spontaneous vasomotion or the structure of the neurovascular unit. This strategy allows the entry of immunoglobulins and viral gene therapy vectors, as well as cargo-laden liposomes. We anticipate this nanotechnology to be useful for tissue regions that are accessible to light or fiberoptic application and to open new avenues for drug screening and therapeutic interventions in the central nervous system.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Vamsidhara Vemireddy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
| | - Qi Cai
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Hejian Xiong
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Peiyuan Kang
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Xiuying Li
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Monica Giannotta
- FIRC Institute of Molecular Oncology Foundation (IFOM), 20139 Milan, Italy
| | - Heather N. Hayenga
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Edward Pan
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
| | - Shashank R. Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United State
| | - Celine Mateo
- Department of Physics, University of California San Diego, La Jolla, California 92093, United State
| | - David Kleinfeld
- Department of Physics, University of California San Diego, La Jolla, California 92093, United State
| | - Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2 D02 PN40, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2 D02 PN40, Ireland
| | - Elisabetta Dejana
- FIRC Institute of Molecular Oncology Foundation (IFOM), 20139 Milan, Italy
| | - Robert Bachoo
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
| | - Zhenpeng Qin
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United State
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, Texas 75080, United State
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United State
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas 75080, United State
| |
Collapse
|
47
|
Xiao X, Huang S, Chen S, Wang Y, Sun Q, Xu X, Li Y. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:367. [PMID: 34794490 PMCID: PMC8600921 DOI: 10.1186/s13046-021-02148-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/20/2021] [Indexed: 02/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has yielded impressive outcomes and transformed treatment algorithms for hematological malignancies. To date, five CAR T-cell products have been approved by the US Food and Drug Administration (FDA). Nevertheless, some significant toxicities pose great challenges to the development of CAR T-cell therapy, most notably cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Understanding the mechanisms underlying these toxicities and establishing prevention and treatment strategies are important. In this review, we summarize the mechanisms underlying CRS and ICANS and provide potential treatment and prevention strategies.
Collapse
Affiliation(s)
- Xinyi Xiao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Shengkang Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Sifei Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Yazhuo Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.,Medical College of Rehabilitation, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Qihang Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, 510005, People's Republic of China.
| |
Collapse
|
48
|
Kaur D, Behl T, Chigurupati S, Sehgal A, Singh S, Sharma N, Badavath VN, Vargas-De-La-Cruz C, Bhatia S, Al-Harrasi A, Dey A, Aleya L, Bungau S. Deciphering the focal role of endostatin in Alzheimer's disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:61998-62011. [PMID: 34561808 DOI: 10.1007/s11356-021-16567-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
Alzheimer's disease (AD) is a paramount chronic neurodegenerative condition that has been affecting elderly people since the 1900s. It causes memory loss, disorientation, and poor mental function. AD is considered to be one of the most serious problems that dementia sufferers face. Despite extensive investigation, the pathological origin of Alzheimer's disease remains a mystery. The amyloid cascade theory and the vascular hypothesis, which stresses the buildup of Aβ plaques, have dominated research into dementia and aging throughout history. However, research into this task failed to yield the long-awaited therapeutic miracle lead for Alzheimer's disease. Perhaps a hypothetical fragility in the context of Alzheimer's disease was regarded as a state distinct from aging in general, as suggested by the angiogenesis hypothesis, which suggests that old age is one state associated with upregulation of angiogenic growth factors, resulting in decreased microcirculation throughout the body. There has also been evidence that by controlling or inhibiting the components involved in the sequence of events that cause angiogenesis, there is a visible progression in AD patients. In Alzheimer's disease, one such antiangiogenic drug is being used.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | | | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Ensenanza e Investigacion en Bacteriologia Alimentaria, Universidad Nacinol Mayor de San Marcos, Lima, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, Peru
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Simona Bungau
- Department of Pharmacy, Faculty of Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
49
|
Schmaier AA, Pajares Hurtado GM, Manickas-Hill ZJ, Sack KD, Chen SM, Bhambhani V, Quadir J, Nath AK, Collier ARY, Ngo D, Barouch DH, Shapiro NI, Gerszten RE, Yu XG, Peters KG, Flaumenhaft R, Parikh SM. Tie2 activation protects against prothrombotic endothelial dysfunction in COVID-19. JCI Insight 2021; 6:e151527. [PMID: 34506304 PMCID: PMC8564889 DOI: 10.1172/jci.insight.151527] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction accompanies the microvascular thrombosis commonly observed in severe COVID-19. Constitutively, the endothelial surface is anticoagulant, a property maintained at least in part via signaling through the Tie2 receptor. During inflammation, the Tie2 antagonist angiopoietin-2 (Angpt-2) is released from endothelial cells and inhibits Tie2, promoting a prothrombotic phenotypic shift. We sought to assess whether severe COVID-19 is associated with procoagulant endothelial dysfunction and alterations in the Tie2/angiopoietin axis. Primary HUVECs treated with plasma from patients with severe COVID-19 upregulated the expression of thromboinflammatory genes, inhibited the expression of antithrombotic genes, and promoted coagulation on the endothelial surface. Pharmacologic activation of Tie2 with the small molecule AKB-9778 reversed the prothrombotic state induced by COVID-19 plasma in primary endothelial cells. Lung autopsies from patients with COVID-19 demonstrated a prothrombotic endothelial signature. Assessment of circulating endothelial markers in a cohort of 98 patients with mild, moderate, or severe COVID-19 revealed endothelial dysfunction indicative of a prothrombotic state. Angpt-2 concentrations rose with increasing disease severity, and the highest levels were associated with worse survival. These data highlight the disruption of Tie2/angiopoietin signaling and procoagulant changes in endothelial cells in severe COVID-19. Our findings provide rationale for current trials of Tie2-activating therapy with AKB-9778 in COVID-19.
Collapse
Affiliation(s)
- Alec A. Schmaier
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Kelsey D. Sack
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Siyu M. Chen
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Victoria Bhambhani
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Juweria Quadir
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Anjali K. Nath
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Debby Ngo
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Dan H. Barouch
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Center for Virology and Vaccine Research, and
| | - Nathan I. Shapiro
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Diseases Division, Brigham and Women’s Hospital and Harvard Medical School, Massachusetts, Boston USA
| | - MGH COVID-19 Collection and Processing Team
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- The MGH COVID-19 Collection and Processing Team is detailed in Supplemental Acknowledgments
| | | | | | - Samir M. Parikh
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology, University of Texas Southwestern, Dallas, Texas, USA
| |
Collapse
|
50
|
Alrafiah A, Alofi E, Almohaya Y, Hamami A, Qadah T, Almaghrabi S, Hakami N, Alrawaili MS, Tayeb HO. Angiogenesis Biomarkers in Ischemic Stroke Patients. J Inflamm Res 2021; 14:4893-4900. [PMID: 34588795 PMCID: PMC8473716 DOI: 10.2147/jir.s331868] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/10/2021] [Indexed: 01/01/2023] Open
Abstract
Introduction Stroke is a global health issue, and ischemic stroke is among the most common strokes affecting many people worldwide. Throughout ischemic stroke, various immune cells counter its effect by releasing cytokines, chemokines, and angiogenic molecules. These molecules can work as potential biomarkers in the diagnosis and monitoring of the progress of ischemic stroke. The current study investigated the use of angiogenic molecules as biomarkers in ischemic stroke patients. Methods The samples were obtained from twenty healthy subjects and nineteen patients with ischemic stroke. Multiplex assay was used to measure the serum levels of angiogenic biomarkers, including endoglin, VEGF-A, endothelin-1, G-CSF, and angiopoietin-2. All data were analyzed using an unpaired Student’s t-test. Correlations between measured parameters were made using Pearson correlations. Results Angiopoietin-2, VEGF-A, endothelin-1, and endoglin levels in stroke patients were significantly higher compared to healthy controls. Nevertheless, G-CSF level showed a non-significant increase in patients compared to controls. The correlation coefficient of measured angiogenic biomarkers among patients showed significant correlations between endoglin, angiopoietin, VEGF-A, and endothelin-1. Discussion The angiogenic factors were significantly increased in patients with ischemic stroke, which may help in the early detection of ischemic stroke and consequently prompt treatment and better prognosis.
Collapse
Affiliation(s)
- Aziza Alrafiah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ebtisam Alofi
- Department of Physiology, Medical School, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yasser Almohaya
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Hamami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Talal Qadah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Safa Almaghrabi
- Department of Physiology, Medical School, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nora Hakami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Moafaq S Alrawaili
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Haythum O Tayeb
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
| |
Collapse
|