1
|
Fernandes MGF, Pernin F, Antel JP, Kennedy TE. From BBB to PPP: Bioenergetic requirements and challenges for oligodendrocytes in health and disease. J Neurochem 2025; 169:e16219. [PMID: 39253904 PMCID: PMC11657931 DOI: 10.1111/jnc.16219] [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: 03/31/2024] [Revised: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Mature myelinating oligodendrocytes, the cells that produce the myelin sheath that insulates axons in the central nervous system, have distinct energetic and metabolic requirements compared to neurons. Neurons require substantial energy to execute action potentials, while the energy needs of oligodendrocytes are directed toward building the lipid-rich components of myelin and supporting neuronal metabolism by transferring glycolytic products to axons as additional fuel. The utilization of energy metabolites in the brain parenchyma is tightly regulated to meet the needs of different cell types. Disruption of the supply of metabolites can lead to stress and oligodendrocyte injury, contributing to various neurological disorders, including some demyelinating diseases. Understanding the physiological properties, structures, and mechanisms involved in oligodendrocyte energy metabolism, as well as the relationship between oligodendrocytes and neighboring cells, is crucial to investigate the underlying pathophysiology caused by metabolic impairment in these disorders. In this review, we describe the particular physiological properties of oligodendrocyte energy metabolism and the response of oligodendrocytes to metabolic stress. We delineate the relationship between oligodendrocytes and other cells in the context of the neurovascular unit, and the regulation of metabolite supply according to energetic needs. We focus on the specific bioenergetic requirements of oligodendrocytes and address the disruption of metabolic energy in demyelinating diseases. We encourage further studies to increase understanding of the significance of metabolic stress on oligodendrocyte injury, to support the development of novel therapeutic approaches for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Milton Guilherme Forestieri Fernandes
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Florian Pernin
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Jack P. Antel
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Timothy E. Kennedy
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
2
|
Dumas A, Goyal N, Mottamal M, Afosah DK, Al-Horani RA. Discovery of a new lead molecule to develop a novel class of human factor XIIa inhibitors. J Thromb Thrombolysis 2024; 57:1308-1314. [PMID: 39487279 DOI: 10.1007/s11239-024-03054-2] [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] [Accepted: 09/09/2024] [Indexed: 11/04/2024]
Abstract
Factor XIIa (FXIIa) is a plasma serine protease within the contact activation pathway. Inhibiting FXIIa could offer a viable therapeutic approach for achieving effective and safer anticoagulation without the bleeding risks that accompany the use of existing anticoagulants. Therefore, we investigated the anticoagulant properties of an amidine-containing molecule (inhibitor 1) to identify a potential lead molecule for subsequent development of FXIIa inhibitors. Results indicated that inhibitor 1 primarily inhibits human FXIIa with an IC50 value of ~30 µM. The inhibitor demonstrated variable selectivity against thrombin, factor IXa, factor Xa, factor XIa, and activated protein C. Michaelis-Menten kinetics indicated that the molecule is an active site inhibitor of FXIIa. Molecular modeling studies revealed that the molecule recognizes residues His57, Asp189, and Ala190 in FXIIa's active site. The inhibitor selectively and concentration-dependently prolonged the clotting time of human plasma under activated partial thromboplastin time assay conditions. The inhibitor did not exhibit significant cytotoxicity in human HEK293 cells and the in silico pharmacokinetics and toxicology data were comparable to known anticoagulants. This study introduces inhibitor 1 as a lead platform for further development as an anticoagulant to provide a more effective and safer approach to preventing and treating thromboembolic diseases.
Collapse
Affiliation(s)
- Anthony Dumas
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Navneet Goyal
- Department of Chemistry, College of Arts and Science, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Madhusoodanan Mottamal
- Department of Chemistry, College of Arts and Science, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Daniel K Afosah
- Department of Medicinal Chemistry, College of Pharmacy, Virginia Commonwealth University, 23298, Richmond, VA, USA
| | - Rami A Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, 70125, USA.
- Drexel Drive, College of Pharmacy, New Orleans, LA, 70125-1089, USA.
| |
Collapse
|
3
|
Cohn DM, Renné T. Targeting factor XIIa for therapeutic interference with hereditary angioedema. J Intern Med 2024; 296:311-326. [PMID: 39331688 DOI: 10.1111/joim.20008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Hereditary angioedema (HAE) is a rare, potentially life-threatening genetic disorder characterized by recurrent attacks of swelling. Local vasodilation and vascular leakage are stimulated by the vasoactive peptide bradykinin, which is excessively produced due to dysregulation of the activated factor XII (FXIIa)-driven kallikrein-kinin system. There is a need for novel treatments for HAE that provide greater efficacy, improved quality of life, minimal adverse effects, and reduced treatment burden over current first-line therapies. FXIIa is emerging as an attractive therapeutic target for interference with HAE attacks. In this review, we draw on preclinical, experimental animal, and in vitro studies, providing an overview on targeting FXIIa as the basis for pharmacologic interference in HAE. We highlight that there is a range of FXIIa inhibitors in development for different therapeutic areas. Of these, garadacimab, an FXIIa-targeted inhibitory monoclonal antibody, is the most advanced and has shown potential as a novel long-term prophylactic treatment for patients with HAE in clinical trials. The evidence from these trials is summarized and discussed, and we propose areas for future research where targeting FXIIa may have therapeutic potential beyond HAE.
Collapse
Affiliation(s)
- Danny M Cohn
- University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Thomas Renné
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
4
|
Zhang Y, Chen Z, Guo J, Wan Q, Zhang Y, Li H, Rao H, Yang J, Xu P, Chen H, Wang M. Factor XII and prekallikrein promote microvascular inflammation and psoriasis in mice. Br J Pharmacol 2024; 181:3760-3778. [PMID: 38872396 DOI: 10.1111/bph.16428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND AND PURPOSE Psoriasis is an autoimmune inflammatory skin disease, featuring microvascular abnormalities and elevated levels of bradykinin. Contact activation of Factor XII can initiate the plasma kallikrein-kinin cascade, producing inflammation and angioedema. The role of Factor XII in psoriasis is unknown. EXPERIMENTAL APPROACH The effects of deficiency of Factor XII or its enzymatic substrate, prekallikrein, were examined in the imiquimod-induced mouse model of psoriasis. Skin microcirculation was assessed using intravital confocal microscopy and laser Doppler flowmeter. A novel antibody blocking Factor XII activation was evaluated for psoriasis prevention. KEY RESULTS Expression of Factor XII was markedly up-regulated in human and mouse psoriatic skin. Genetic deletion of Factor XII or prekallikrein, attenuated imiquimod-induced psoriatic lesions in mice. Psoriatic induction increased skin microvascular blood perfusion, causing vasodilation, hyperpermeability and angiogenesis. It also promoted neutrophil-vascular interaction, inflammatory cytokine release and enhanced Factor XII / prekallikrein enzymatic activity with elevated bradykinin. Factor XII or prekallikrein deficiency ameliorated these microvascular abnormalities and abolished bradykinin increase. Antagonism of bradykinin B2 receptors reproduced the microvascular protection of Factor XII / prekallikrein deficiency, attenuated psoriatic lesions, and prevented protection by Factor XII / prekallikrein deficiency against psoriasis. Furthermore, treatment of mice with Factor XII antibody alleviated experimentally induced psoriasis and suppressed microvascular inflammation. CONCLUSION AND IMPLICATIONS Activation of Factor XII promoted psoriasis via prekallikrein-dependent formation of bradykinin, which critically mediated psoriatic microvascular inflammation. Inhibition of contact activation represents a novel therapeutic strategy for psoriasis.
Collapse
Affiliation(s)
- Yurong Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zengrong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyan Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Life Science, Zhejiang Normal University, Jinhua City, China
| | - Qing Wan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingjie Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huihui Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haojie Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianfeng Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengfei 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, China
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Xu P, Zhang Y, Guo J, Li H, Konrath S, Zhou P, Cai L, Rao H, Chen H, Lin J, Cui Z, Ji B, Wang J, Li N, Liu DP, Renné T, Wang M. A single-domain antibody targeting factor XII inhibits both thrombosis and inflammation. Nat Commun 2024; 15:7898. [PMID: 39266545 PMCID: PMC11393108 DOI: 10.1038/s41467-024-51745-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Factor XII (FXII) is the zymogen of the plasma protease FXIIa that activates the intrinsic coagulation pathway and the kallikrein kinin-system. The role of FXII in inflammation has been obscure. Here, we report a single-domain antibody (nanobody, Nb) fused to the Fc region of a human immunoglobulin (Nb-Fc) that recognizes FXII in a conformation-dependent manner and interferes with FXIIa formation. Nb-Fc treatment inhibited arterial thrombosis in male mice without affecting hemostasis. In a mouse model of extracorporeal membrane oxygenation (ECMO), FXII inhibition or knockout reduced thrombus deposition on oxygenator membranes and systemic microvascular thrombi. ECMO increased circulating levels of D-dimer, alkaline phosphatase, creatinine and TNF-α and triggered microvascular neutrophil adherence, platelet aggregation and their interaction, which were substantially attenuated by FXII blockade. Both Nb-Fc treatment and FXII knockout markedly ameliorated immune complex-induced local vasculitis and anti-neutrophil cytoplasmic antibody-induced systemic vasculitis, consistent with selectively suppressed neutrophil migration. In human blood microfluidic analysis, Nb-Fc treatment prevented collagen-induced fibrin deposition and neutrophil adhesion/activation. Thus, FXII is an important mediator of inflammatory responses in vasculitis and ECMO, and Nb-Fc provides a promising approach to alleviate thrombo-inflammatory disorders.
Collapse
Affiliation(s)
- Pengfei 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, China
| | - Yingjie Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyan Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Life Science, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Huihui Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Peng Zhou
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Liming Cai
- Department of Cardiopulmonary Bypass, State Key Laboratory of Cardiovascular Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haojie Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhao Cui
- Renal Division, Peking University First Hospital, Beijing, China
| | - Bingyang Ji
- Department of Cardiopulmonary Bypass, State Key Laboratory of Cardiovascular Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nailin Li
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institute, Stockholm, Sweden
| | - De-Pei Liu
- Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- National Health Commission Cardiovascular Disease Regenerative Medicine Research Key Laboratory, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
6
|
Elwakiel A, Gupta D, Rana R, Manoharan J, Al-Dabet MM, Ambreen S, Fatima S, Zimmermann S, Mathew A, Li Z, Singh K, Gupta A, Pal S, Sulaj A, Kopf S, Schwab C, Baber R, Geffers R, Götze T, Alo B, Lamers C, Kluge P, Kuenze G, Kohli S, Renné T, Shahzad K, Isermann B. Factor XII signaling via uPAR-integrin β1 axis promotes tubular senescence in diabetic kidney disease. Nat Commun 2024; 15:7963. [PMID: 39261453 PMCID: PMC11390906 DOI: 10.1038/s41467-024-52214-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Coagulation factor XII (FXII) conveys various functions as an active protease that promotes thrombosis and inflammation, and as a zymogen via surface receptors like urokinase-type plasminogen activator receptor (uPAR). While plasma levels of FXII are increased in diabetes mellitus and diabetic kidney disease (DKD), a pathogenic role of FXII in DKD remains unknown. Here we show that FXII is locally expressed in kidney tubular cells and that urinary FXII correlates with kidney dysfunction in DKD patients. F12-deficient mice (F12-/-) are protected from hyperglycemia-induced kidney injury. Mechanistically, FXII interacts with uPAR on tubular cells promoting integrin β1-dependent signaling. This signaling axis induces oxidative stress, persistent DNA damage and senescence. Blocking uPAR or integrin β1 ameliorates FXII-induced tubular cell injury. Our findings demonstrate that FXII-uPAR-integrin β1 signaling on tubular cells drives senescence. These findings imply previously undescribed diagnostic and therapeutic approaches to detect or treat DKD and possibly other senescence-associated diseases.
Collapse
Affiliation(s)
- Ahmed Elwakiel
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany.
| | - Dheerendra Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Jayakumar Manoharan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Moh'd Mohanad Al-Dabet
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
- Department of Medical Laboratory Sciences, School of Science, University of Jordan, Amman, Jordan
| | - Saira Ambreen
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Sameen Fatima
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Silke Zimmermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Zhiyang Li
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Kunal Singh
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Anubhuti Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Surinder Pal
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Alba Sulaj
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), University of Heidelberg, Heidelberg, Germany
| | - Stefan Kopf
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), University of Heidelberg, Heidelberg, Germany
| | - Constantin Schwab
- Institute of pathology, University of Heidelberg, Heidelberg, Germany
| | - Ronny Baber
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
- Leipzig Medical Biobank, Leipzig University, Leipzig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tom Götze
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Bekas Alo
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Christina Lamers
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Paul Kluge
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Georg Kuenze
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University, Leipzig, Germany
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Khurrum Shahzad
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
7
|
Frunt R, El Otmani H, Smits S, Clark CC, Maas C. Factor XII contact activation can be prevented by targeting 2 unique patches in its epidermal growth factor-like 1 domain with a nanobody. J Thromb Haemost 2024; 22:2562-2575. [PMID: 38897387 DOI: 10.1016/j.jtha.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Factor (F)XII triggers contact activation by binding to foreign surfaces, with the epidermal growth factor-like 1 (EGF-1) domain being the primary binding site. Blocking FXII surface-binding might hold therapeutic value to prevent medical device-induced thrombosis. OBJECTIVES To unravel and prevent EGF-1-mediated FXII surface-binding with a variable domain of heavy chain-only antibody (VHH). METHODS FXII variants with glutamine substitutions of 2 positively charged amino acid patches within the EGF-1 domain were created. Their role in FXII contact activation was assessed using kaolin pull-down experiments, amidolytic activity assays, and clotting assays. FXII EGF-1 domain-specific VHHs were raised to inhibit EGF-1-mediated FXII contact activation while preserving quiescence. RESULTS Two unique, positively charged patches in the EGF-1 domain were identified (upstream, 73K74K76K78H81K82H; downstream, 87K113K). Neutralizing the charge of both patches led to a 99% reduction in FXII kaolin binding, subsequent decrease in autoactivation of 94%, and prolongation of clot formation in activated partial thromboplastin time assays from 36 (±2) to 223 (±13) seconds. Three FXII EGF-1-specific VHHs were developed that are capable of inhibiting kaolin binding and subsequent contact system activation in plasma. The most effective VHH "F2" binds the positively charged patches and thereby dose-dependently extends activated partial thromboplastin time clotting times from 29 (±2) to 43 (±3) seconds without disrupting FXII quiescence. CONCLUSION The 2 unique, positively charged patches in FXII EGF-1 cooperatively mediate FXII surface-binding, making both patches crucial for contact activation. Targeting these with FXII EGF-1-specific VHHs can exclusively decrease FXII surface-binding and subsequent contact activation, while preserving zymogen quiescence. These patches thus have potential as druggable targets in preventing medical device-induced thrombosis.
Collapse
Affiliation(s)
- Rowan Frunt
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Hinde El Otmani
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Simone Smits
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Chantal C Clark
- Center for Benign Hematology, Thrombosis and Hemostasis - Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Coen Maas
- Central Diagnostic Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
8
|
Jacobs BM, Vickaryous N, Giovannoni G, Proitsi P, Waters S, Dobson R. Plasma proteomic profiles of UK Biobank participants with multiple sclerosis. Ann Clin Transl Neurol 2024; 11:698-709. [PMID: 38282238 PMCID: PMC10963287 DOI: 10.1002/acn3.51990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
OBJECTIVE We aimed to describe plasma protein biomarkers of multiple sclerosis risk and to explore protein biomarkers of disease severity using radiological outcome measures. METHODS Multiple sclerosis cases and controls were identified in UK Biobank, a longitudinal cohort study of ~500,000 British adults. Plasma proteins were assayed in ~50,000 UK Biobank participants using the Olink proximity extension assay. We performed case-control association testing to examine the association between 2911 proteins and multiple sclerosis, using linear models adjusted for confounding covariates. Associations with radiological lesion burden and brain volume were determined in a subset of the cohort with available magnetic resonance imaging, using normalized T2-hyperintensity volume or whole brain volume as the outcome measure. RESULTS In total, 407 prevalent multiple sclerosis cases and 39,979 healthy controls were included. We discovered 72 proteins associated with multiple sclerosis at a Bonferroni-adjusted p value of 0.05, including established markers such as neurofilament light chain and glial fibrillary acidic protein. We observed a decrease in plasma Granzyme A, a marker of T cell and NK cell degranulation, which was specific to multiple sclerosis. Higher levels of plasma proteins involved in coagulation were associated with lower T2 lesion burden and preserved brain volume. INTERPRETATION We report the largest plasma proteomic screen of multiple sclerosis, replicating important known associations and suggesting novel markers, such as the reduction in granzyme A. While these findings require external validation, they demonstrate the power of biobank-scale datasets for discovering new biomarkers for multiple sclerosis.
Collapse
Affiliation(s)
- Benjamin M. Jacobs
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
- Department of NeurologyRoyal London HospitalLondonUK
| | - Nicola Vickaryous
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Gavin Giovannoni
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
- Department of NeurologyRoyal London HospitalLondonUK
| | - Petroula Proitsi
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Sheena Waters
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| | - Ruth Dobson
- Centre for Preventive Neurology, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
- Department of NeurologyRoyal London HospitalLondonUK
| |
Collapse
|
9
|
Lyu J, Chen H, Luo J, Lin S, Yang G, Zhou M, Tao J. Shape memory and hemostatic silk-laponite scaffold for alveolar bone regeneration after tooth extraction trauma. Int J Biol Macromol 2024; 260:129454. [PMID: 38237836 DOI: 10.1016/j.ijbiomac.2024.129454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
Persistent bleeding and the absence of alveolar bone stress following tooth loss can hinder socket healing, complicating future dental implant procedures, and potentially leading to neighboring tooth instability. Therefore, developing materials that promote alveolar bone regeneration and possess both hemostatic and osteogenic properties is crucial for preserving the extraction sites. This study introduces a silk-based laponite composite scaffold material with proficient hemostatic and osteogenic functions, and excellent shape-memory properties for efficient extraction- site filling. In vitro studies research demonstrated that the scaffold's inherent negative charge of the scaffold significantly enhanced blood coagulation and thrombin generation. Moreover, its porous structure and slightly rough inner surface promoted blood cell adhesion and, improved the hemostatic performance. Furthermore, the scaffold facilitated stem cell osteogenic differentiation by activating the TRPM7 channel through the released of magnesium ions. In vivo tests using rat models confirmed its effectiveness in promoting coagulation and mandibular regeneration. Thus, this study proposes a promising approach for post-extraction alveolar bone regenerative repair. The composite scaffold material, with its hemostatic and osteogenic capabilities and shape-memory features, can potentially enhance dental implant success and overall oral health.
Collapse
Affiliation(s)
- Jiaxuan Lyu
- Department of General Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Hongyan Chen
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Jiaxin Luo
- Department of Dental Implantology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, No. 195 Dongfengwest Road, Guangzhou 510160, China
| | - Sihan Lin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Mingliang Zhou
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China.
| | - Jiang Tao
- Department of General Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639 Zhizaoju Road, Shanghai 200011, China.
| |
Collapse
|
10
|
Taskin B, Kohs TC, Shatzel JJ, Puy C, McCarty OJ. Factor XI as a therapeutic target in neuroinflammatory disease. Curr Opin Hematol 2024; 31:32-38. [PMID: 37694771 PMCID: PMC10843631 DOI: 10.1097/moh.0000000000000787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the pathophysiology and potential therapeutic options for treatment of multiple sclerosis, a common neuronal demyelinating disorder affecting 2.2 million people worldwide. As an autoimmune disorder, multiple sclerosis is associated with neuroinflammation and increased permeability of the blood-brain barrier (BBB), although the cause linking multiple sclerosis with compromised barrier function remains ill-defined. It has been previously shown that coagulation factors, including thrombin and fibrin, exacerbate the inflammatory processes and permeability of the BBB. RECENT FINDINGS Increased levels of the coagulation factor (F) XII have been found in patients presenting with relapsing-remitting multiple sclerosis, with a deleterious role for FXII being validated in murine model of multiple sclerosis, experimental autoimmune encephalitis (EAE). Recent work has uncovered a role for the major substrate activated by FXII and thrombin, FXI, in the disorder of EAE. The study found that pharmacological targeting of FXI decreased clinical symptoms, lymphocyte invasion, and white matter destruction in a multiple sclerosis model. SUMMARY This review emphasizes the role of FXII and FXI in regulating barrier function and the immune response in neuroinflammation. These new findings broaden the potential for therapeutic utility of FXI inhibitors beyond thrombosis to include neuroinflammatory diseases associated with compromised BBB function, including multiple sclerosis.
Collapse
Affiliation(s)
- Berk Taskin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
11
|
Zhao J, Zhang X, Li Y, Yu J, Chen Z, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Hao Y, Zong J, Xia C, Xia J, Wu J. Interorgan communication with the liver: novel mechanisms and therapeutic targets. Front Immunol 2023; 14:1314123. [PMID: 38155961 PMCID: PMC10754533 DOI: 10.3389/fimmu.2023.1314123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
The liver is a multifunctional organ that plays crucial roles in numerous physiological processes, such as production of bile and proteins for blood plasma, regulation of blood levels of amino acids, processing of hemoglobin, clearance of metabolic waste, maintenance of glucose, etc. Therefore, the liver is essential for the homeostasis of organisms. With the development of research on the liver, there is growing concern about its effect on immune cells of innate and adaptive immunity. For example, the liver regulates the proliferation, differentiation, and effector functions of immune cells through various secreted proteins (also known as "hepatokines"). As a result, the liver is identified as an important regulator of the immune system. Furthermore, many diseases resulting from immune disorders are thought to be related to the dysfunction of the liver, including systemic lupus erythematosus, multiple sclerosis, and heart failure. Thus, the liver plays a role in remote immune regulation and is intricately linked with systemic immunity. This review provides a comprehensive overview of the liver remote regulation of the body's innate and adaptive immunity regarding to main areas: immune-related molecules secreted by the liver and the liver-resident cells. Additionally, we assessed the influence of the liver on various facets of systemic immune-related diseases, offering insights into the clinical application of target therapies for liver immune regulation, as well as future developmental trends.
Collapse
Affiliation(s)
- Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
12
|
Padilla S, Prado R, Anitua E. An evolutionary history of F12 gene: Emergence, loss, and vulnerability with the environment as a driver. Bioessays 2023; 45:e2300077. [PMID: 37750435 DOI: 10.1002/bies.202300077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
In the context of macroevolutionary transitions, environmental changes prompted vertebrates already bearing genetic variations to undergo gradual adaptations resulting in profound anatomical, physiological, and behavioral adaptations. The emergence of new genes led to the genetic variation essential in metazoan evolution, just as was gene loss, both sources of genetic variation resulting in adaptive phenotypic diversity. In this context, F12-coding protein with defense and hemostatic roles emerged some 425 Mya, and it might have contributed in aquatic vertebrates to the transition from water-to-land. Conversely, the F12 loss in marine, air-breathing mammals like cetaceans has been associated with phenotypic adaptations in some terrestrial mammals in their transition to aquatic lifestyle. More recently, the advent of technological innovations in western lifestyle with blood-contacting devices and harmful environmental nanoparticles, has unfolded new roles of FXII. Environment operates as either a positive or a relaxed selective pressure on genes, and consequently genes are selected or lost. FXII, an old dog facing environmental novelties can learn new tricks and teach us new therapeutic avenues.
Collapse
Affiliation(s)
- Sabino Padilla
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Roberto Prado
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Eduardo Anitua
- BTI-Biotechnology Institute ImasD, Vitoria, Spain
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain
- University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| |
Collapse
|
13
|
Chen SS, Luo HJ, Li H, Zhang H, Li YY, Li Y, Wang W, Ding HL. Elevated plasma FXII is associated with disease activity in ANCA-associated vasculitis: A Retrospective Cross-Sectional Study in Western China. Int Immunopharmacol 2023; 125:111067. [PMID: 37866310 DOI: 10.1016/j.intimp.2023.111067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
This study examined whether plasma FXII levels reflect disease activity in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). Plasma FXII levels were detected by ELISA in 127 patients with AAV, and their associations with disease activity and plasma myeloperoxidase (MPO)-ANCA titre were examined. Immunofluorescent co-staining of FXII and neutrophils was performed on the renal tissues of patients with AAV. MPO expression in renal biopsy tissues was determined by immunohistochemical staining. The association between plasma FXII levels and histological activity was assessed in 82 patients who underwent kidney biopsy. Plasma FXII levels were considerably increased in patients with clinically active AAV compared to those in clinical remission and healthy individuals. Plasma FXII levels correlated positively with creatinine (r = 0.377), CRP (r = 0.222), urine red blood cell (r = 0.203), serum MPO-ANCA titer (r = 0.353), white blood cell (r = 0.194), percentage of glomeruli with crescents (P = 0.001), capillary breaks (P = 0.001), interstitial inflammation (P < 0.001) and fibrinoid necrosis (p < 0.001) on kidney biopsy. The plasma FXII optimal cut-off value for evaluating AAV activity was 24.5 μg/mL (sensitivity = 0.81, specificity = 0.82, P = 0.0001), which was superior to that achieved using conventional serologic biomarkers. Co-expression of FXII and neutrophils was higher, with increased MPO expression, in renal tissue with pathologically active AAV than that observed in pathologically inactive tissues. In conclusion, elevated plasma FXII levels reflect AAV clinical and histologic activity, and can serve as markers of active AAV.
Collapse
Affiliation(s)
- Sha-Sha Chen
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Hao-Jun Luo
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China; Department of Palliative Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Huan Li
- Wenjiang District People's Hospital of Chengdu, Wenjiang Hospital of Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan, China
| | - Hong Zhang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Yuan-Yuan Li
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Yi Li
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Wei Wang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Han-Lu Ding
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.
| |
Collapse
|
14
|
Sharp RC, Guenther DT, Farrer MJ. Experimental procedures for flow cytometry of wild-type mouse brain: a systematic review. Front Immunol 2023; 14:1281705. [PMID: 38022545 PMCID: PMC10646240 DOI: 10.3389/fimmu.2023.1281705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The aim of this study was to systematically review the neuroimmunology literature to determine the average immune cell counts reported by flow cytometry in wild-type (WT) homogenized mouse brains. Background Mouse models of gene dysfunction are widely used to study age-associated neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. The importance of the neuroimmune system in these multifactorial disorders has become increasingly evident, and methods to quantify resident and infiltrating immune cells in the brain, including flow cytometry, are necessary. However, there appears to be no consensus on the best approach to perform flow cytometry or quantify/report immune cell counts. The development of more standardized methods would accelerate neuroimmune discovery and validation by meta-analysis. Methods There has not yet been a systematic review of 'neuroimmunology' by 'flow cytometry' via examination of the PROSPERO registry. A protocol for a systematic review was subsequently based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) using the Studies, Data, Methods, and Outcomes (SDMO) criteria. Literature searches were conducted in the Google Scholar and PubMed databases. From that search, 900 candidate studies were identified, and 437 studies were assessed for eligibility based on formal exclusion criteria. Results Out of the 437 studies reviewed, 58 were eligible for inclusion and comparative analysis. Each study assessed immune cell subsets within homogenized mouse brains and used flow cytometry. Nonetheless, there was considerable variability in the methods, data analysis, reporting, and results. Descriptive statistics have been presented on the study designs and results, including medians with interquartile ranges (IQRs) and overall means with standard deviations (SD) for specific immune cell counts and their relative proportions, within and between studies. A total of 58 studies reported the most abundant immune cells within the brains were TMEM119+ microglia, bulk CD4+ T cells, and bulk CD8+ T cells. Conclusion Experiments to conduct and report flow cytometry data, derived from WT homogenized mouse brains, would benefit from a more standardized approach. While within-study comparisons are valid, the variability in methods of counting of immune cell populations is too broad for meta-analysis. The inclusion of a minimal protocol with more detailed methods, controls, and standards could enable this nascent field to compare results across studies.
Collapse
Affiliation(s)
| | | | - Matthew J. Farrer
- Department of Neurology, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Kohs TCL, Fallon ME, Oseas EC, Healy LD, Tucker EI, Gailani D, McCarty OJT, Vandenbark AA, Offner H, Verbout NG. Pharmacological targeting of coagulation factor XI attenuates experimental autoimmune encephalomyelitis in mice. Metab Brain Dis 2023; 38:2383-2391. [PMID: 37341855 PMCID: PMC10530106 DOI: 10.1007/s11011-023-01251-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
Multiple sclerosis (MS) is the most common causes of non-traumatic disability in young adults worldwide. MS pathophysiologies include the formation of inflammatory lesions, axonal damage and demyelination, and blood brain barrier (BBB) disruption. Coagulation proteins, including factor (F)XII, can serve as important mediators of the adaptive immune response during neuroinflammation. Indeed, plasma FXII levels are increased during relapse in relapsing-remitting MS patients, and previous studies showed that reducing FXII levels was protective in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). Our objective was to determine if pharmacological targeting of FXI, a major substrate of activated FXII (FXIIa), improves neurological function and attenuates CNS damage in the setting of EAE. EAE was induced in male mice using murine myelin oligodendrocyte glycoprotein peptides combined with heat-inactivated Mycobacterium tuberculosis and pertussis toxin. Upon onset of symptoms, mice were treated every other day intravenously with anti-FXI antibody, 14E11, or saline. Disease scores were recorded daily until euthanasia for ex vivo analyses of inflammation. Compared to the vehicle control, 14E11 treatment reduced the clinical severity of EAE and total mononuclear cells, including CD11b+CD45high macrophage/microglia and CD4+ T cell numbers in brain. Following pharmacological targeting of FXI, BBB disruption was reduced, as measured by decreased axonal damage and fibrin(ogen) accumulation in the spinal cord. These data demonstrate that pharmacological inhibition of FXI reduces disease severity, immune cell migration, axonal damage, and BBB disruption in mice with EAE. Thus, therapeutic agents targeting FXI and FXII may provide a useful approach for treating autoimmune and neurologic disorders.
Collapse
Affiliation(s)
- Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA.
| | - Meghan E Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Ethan C Oseas
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Laura D Healy
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Erik I Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
- Aronora, Inc., Portland, OR, USA
| | - David Gailani
- Department of Pathology and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
- Veterans Affairs Portland Health Care System, Portland, OR, USA
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Norah G Verbout
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S. Bond Avenue, Portland, OR, 97239, USA
- Aronora, Inc., Portland, OR, USA
| |
Collapse
|
16
|
Wahab R, Hasan MM, Azam Z, Grippo PJ, Al-Hilal TA. The role of coagulome in the tumor immune microenvironment. Adv Drug Deliv Rev 2023; 200:115027. [PMID: 37517779 PMCID: PMC11099942 DOI: 10.1016/j.addr.2023.115027] [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: 01/31/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
The rising incidence and persistent thrombosis in multiple cancers including those that are immunosuppressive highlight the need for understanding the tumor coagulome system and its role beyond hemostatic complications. Immunotherapy has shown significant benefits in solid organ tumors but has been disappointing in the treatment of hypercoagulable cancers, such as glioblastoma and pancreatic ductal adenocarcinomas. Thus, targeting thrombosis to prevent immunosuppression seems a clinically viable approach in cancer treatment. Hypercoagulable tumors often develop fibrin clots within the tumor microenvironment (TME) that dictates the biophysical characteristics of the tumor tissue. The application of systems biology and single-cell approaches highlight the potential role of coagulome or thrombocytosis in shaping the tumor immune microenvironment (TIME). In-depth knowledge of the tumor coagulome would provide unprecedented opportunities to better predict the hemostatic complications, explore how thrombotic stroma modulates tumor immunity, reexamine the significance of clinical biomarkers, and enable steering the stromal versus systemic immune response for boosting the effectiveness of immune checkpoint inhibitors in cancer treatment. We focus on the role of coagulation factors in priming a suppressive TIME and the huge potential of existing anticoagulant drugs in the clinical settings of cancer immunotherapy.
Collapse
Affiliation(s)
- Riajul Wahab
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Md Mahedi Hasan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Zulfikar Azam
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA.
| |
Collapse
|
17
|
Jia X, Hua C, Yang F, Li X, Zhao P, Zhou F, Lu Y, Liang H, Xing M, Lyu G. Hydrophobic aerogel-modified hemostatic gauze with thermal management performance. Bioact Mater 2023; 26:142-158. [PMID: 36911208 PMCID: PMC9996136 DOI: 10.1016/j.bioactmat.2023.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Current hemostatic agents or dressings are not efficient under extremely hot and cold environments due to deterioration of active ingredients, water evaporation and ice crystal growth. To address these challenges, we engineered a biocompatible hemostatic system with thermoregulatory properties for harsh conditions by combining the asymmetric wetting nano-silica aerogel coated-gauze (AWNSA@G) with a layer-by-layer (LBL) structure. Our AWNSA@G was a dressing with a tunable wettability prepared by spraying the hydrophobic nano-silica aerogel onto the gauze from different distances. The hemostatic time and blood loss of the AWNSA@G were 5.1 and 6.9 times lower than normal gauze in rat's injured femoral artery model. Moreover, the modified gauze was torn off after hemostasis without rebleeding, approximately 23.8 times of peak peeling force lower than normal gauze. For the LBL structure, consisting of the nano-silica aerogel layer and a n-octadecane phase change material layer, in both hot (70 °C) and cold (-27 °C) environments, exhibited dual-functional thermal management and maintained a stable internal temperature. We further verified our composite presented superior blood coagulation effect in extreme environments due to the LBL structure, the pro-coagulant properties of nano-silica aerogel and unidirectional fluid pumping of AWNSA@G. Our work, therefore, shows great hemostasis potential under normal and extreme temperature environments.
Collapse
Affiliation(s)
- Xiaoli Jia
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Chao Hua
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Medical School of Nantong University, Nantong, 226019, China
| | - Fengbo Yang
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xiaoxiao Li
- Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Peng Zhao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Feifan Zhou
- Medical School of Nantong University, Nantong, 226019, China
| | - Yichi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Hao Liang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Guozhong Lyu
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.,Medical School of Nantong University, Nantong, 226019, China.,Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
18
|
Schwab JM, Haider C, Kopp MA, Zrzavy T, Endmayr V, Ricken G, Kubista H, Haider T, Liebscher T, Lübstorf T, Blex C, Serdani-Neuhaus L, Curt A, Cinelli P, Scivoletto G, Fehlings MG, May C, Guntermann A, Marcus K, Meisel C, Dirnagl U, Martus P, Prüss H, Popovich PG, Lassmann H, Höftberger R. Lesional Antibody Synthesis and Complement Deposition Associate With De Novo Antineuronal Antibody Synthesis After Spinal Cord Injury. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200099. [PMID: 37019668 PMCID: PMC10075523 DOI: 10.1212/nxi.0000000000200099] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 01/06/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND AND OBJECTIVES Spinal cord injury (SCI) disrupts the fine-balanced interaction between the CNS and immune system and can cause maladaptive aberrant immune responses. The study examines emerging autoantibody synthesis after SCI with binding to conformational spinal cord epitopes and surface peptides located on the intact neuronal membrane. METHODS This is a prospective longitudinal cohort study conducted in acute care and inpatient rehabilitation centers in conjunction with a neuropathologic case-control study in archival tissue samples ranging from acute injury (baseline) to several months thereafter (follow-up). In the cohort study, serum autoantibody binding was examined in a blinded manner using tissue-based assays (TBAs) and dorsal root ganglia (DRG) neuronal cultures. Groups with traumatic motor complete SCI vs motor incomplete SCI vs isolated vertebral fracture without SCI (controls) were compared. In the neuropathologic study, B cell infiltration and antibody synthesis at the spinal lesion site were examined by comparing SCI with neuropathologically unaltered cord tissue. In addition, the CSF in an individual patient was explored. RESULTS Emerging autoantibody binding in both TBA and DRG assessments was restricted to an SCI patient subpopulation only (16%, 9/55 sera) while being absent in vertebral fracture controls (0%, 0/19 sera). Autoantibody binding to the spinal cord characteristically detected the substantia gelatinosa, a less-myelinated region of high synaptic density involved in sensory-motor integration and pain processing. Autoantibody binding was most frequent after motor complete SCI (grade American Spinal Injury Association impairment scale A/B, 22%, 8/37 sera) and was associated with neuropathic pain medication. In conjunction, the neuropathologic study demonstrated lesional spinal infiltration of B cells (CD20, CD79a) in 27% (6/22) of patients with SCI, the presence of plasma cells (CD138) in 9% (2/22). IgG and IgM antibody syntheses colocalized to areas of activated complement (C9neo) deposition. Longitudinal CSF analysis of an additional single patient demonstrated de novo (IgM) intrathecal antibody synthesis emerging with late reopening of the blood-spinal cord barrier. DISCUSSION This study provides immunologic, neurobiological, and neuropathologic proof-of-principle for an antibody-mediated autoimmunity response emerging approximately 3 weeks after SCI in a patient subpopulation with a high demand of neuropathic pain medication. Emerging autoimmunity directed against specific spinal cord and neuronal epitopes suggests the existence of paratraumatic CNS autoimmune syndromes.
Collapse
Affiliation(s)
- Jan M Schwab
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria.
| | - Carmen Haider
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Marcel A Kopp
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Tobias Zrzavy
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Verena Endmayr
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Gerda Ricken
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Helmut Kubista
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Thomas Haider
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Thomas Liebscher
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Tom Lübstorf
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Christian Blex
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Leonarda Serdani-Neuhaus
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Armin Curt
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Paolo Cinelli
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Giorgio Scivoletto
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Michael G Fehlings
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Caroline May
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Annika Guntermann
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Katrin Marcus
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Christian Meisel
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Ulrich Dirnagl
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Peter Martus
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Harald Prüss
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Phillip G Popovich
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Hans Lassmann
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria
| | - Romana Höftberger
- From the The Belford Center for Spinal Cord Injury (J.M.S., P.G.P.), The Ohio State University, Wexner Medical Center, Columbus; Departments of Neurology (J.M.S.), Physical Medicine and Rehabilitation, and Neurosciences, The Ohio State University, Columbus; Department of Neurology and Experimental Neurology (J.M.S., M.A.K., T. Liebscher, T. Lübstorf, C.B., L.S.-N., U.D., H.P.), Spinal Cord Injury Research (Neuroparaplegiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Division of Neuropathology and Neurochemistry (C.H., V.E., G.R., R.H.), Department of Neurology, Medical University of Vienna, Austria; Department of Neurology (T.Z.), Medical University of Vienna, Austria; Department of Neurophysiology and Neuropharmacology (Center for Physiology and Pharmacology) (H.K.), Medical University of Vienna, Austria; Department of Orthopaedics and Trauma Surgery (T.H.), Medical University of Vienna, Austria; Treatment Centre for Spinal Cord Injuries (Thomas Liebscher), BG Hospital Unfallkrankenhaus Berlin, Germany; Spinal Cord Injury Center (A.C.), Balgrist University Hospital, Zurich, Switzerland; Division of Trauma Surgery (P.C.), University Hospital Zürich, Switzerland; IRCCS Fondazione S. Lucia (G.S.), Spinal Cord Unit, Rome, Italy; Division of Neurosurgery and Spine Program (M.G.F.), University of Toronto, ON, Canada; Ruhr-University Bochum (C. May, A.G., K.M.), Center for Protein Diagnostics (PRODI), Medical Proteome Center, Universitätsstraße 150, Bochum, Germany; Institute of Medical Immunology (C. Meisel), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Immunology (C. Meisel), Labor Berlin-Charité Vivantes GmbH, Germany; Berlin Institute of Health (U.D.), QUEST-Center for Transforming Biomedical Research, Germany; Department of Clinical Epidemiology and Applied Biostatistics (P.M.), Eberhard Karls Universität Tübingen, Germany; Department of Neurosciences (P.G.P.), The Ohio State University, Columbus; and Center for Brain Research (H.L.), Medical University of Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health (C.H., T.Z., V.E., G.R., R.H.), Medical University of Vienna, Austria.
| |
Collapse
|
19
|
A Preclinical Investigation on the Role of IgG Antibodies against Coagulant Components in Multiple Sclerosis. Biomedicines 2023; 11:biomedicines11030906. [PMID: 36979885 PMCID: PMC10046059 DOI: 10.3390/biomedicines11030906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
The coagulation-inflammation interplay has recently been identified as a critical risk factor in the early onset of multiple sclerosis (MS), and antibodies against coagulation components have been recognized as contributing factors to thrombotic and inflammatory signaling pathways in diseases with overlapping symptoms to MS, paving the way for further research into their effects on MS pathology. The current study aimed to enlighten the role of IgG antibodies against coagulation components by performing a preclinical study, analyzing the astrocytic activation by purified IgG antibodies derived from 15 MS patients, and assessing their possible pro-inflammatory effects using a bead-based multiplexed immunoassay system. The results were compared with those obtained following astrocyte treatment with samples from 14 age- and gender-matched healthy donors, negative for IgG antibody presence. Serum samples collected from 167 MS patients and 40 age- and gender-matched controls were also analyzed for pro- and anti-inflammatory factors. According to our results, astrocytic activation in response to IgG treatment caused an upregulation of various pro-inflammatory factors, including cytokines, chemokines, and interleukins. Conversely, in serum samples from patients and controls, the pro-inflammatory factors did not differ significantly; medication may lower the levels in patients. Our findings suggest that antibodies may function as effectors in neuroinflammation and serve as targets for new treatments that eventually benefit novel therapeutic approaches.
Collapse
|
20
|
Al-Horani RA. 6-(Arylaminomethyl) Isoquinolines as Enzyme Inhibitors and Their Preparation: A Patent Highlight of Factor XIIa Inhibitors. Cardiovasc Hematol Agents Med Chem 2023; 21:243-249. [PMID: 36703578 PMCID: PMC10501477 DOI: 10.2174/1871525721666230126114224] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/07/2022] [Accepted: 11/16/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans LA 70125, USA
| |
Collapse
|
21
|
Al-Horani RA, Afosah DK, Mottamal M. Triazol-1-yl Benzamides Promote Anticoagulant Activity via Inhibition of Factor XIIa. Cardiovasc Hematol Agents Med Chem 2023; 21:108-119. [PMID: 36321236 PMCID: PMC10249145 DOI: 10.2174/1871525721666221031141323] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/13/2022] [Accepted: 10/03/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Human factor XIIa (FXIIa) is a plasma serine protease that plays a significant role in several physiological and pathological processes. Animal models have revealed an important contribution of FXIIa to thromboembolic diseases. Remarkably, animals and patients with FXII deficiency appear to have normal hemostasis. Thus, FXIIa inhibition may serve as a promising therapeutic strategy to attain safer and more effective anticoagulation. Very few small molecule inhibitors of FXIIa have been reported. We synthesized and investigated a focused library of triazol-1-yl benzamide derivatives for FXIIa inhibition. METHODS We chemically synthesized, characterized, and investigated a focused library of triazol- 1-yl benzamide derivatives for FXIIa inhibition. Using a standardized chromogenic substrate hydrolysis assay, the derivatives were evaluated for inhibiting human FXIIa. Their selectivity over other clotting factors was also evaluated using the corresponding substrate hydrolysis assays. The best inhibitor affinity to FXIIa was also determined using fluorescence spectroscopy. Effects on the clotting times (prothrombin time (PT) and activated partial thromboplastin time (APTT)) of human plasma were also studied. RESULTS We identified a specific derivative (1) as the most potent inhibitor in this series. The inhibitor exhibited nanomolar binding affinity to FXIIa. It also exhibited significant selectivity against several serine proteases. It also selectively doubled the activated partial thromboplastin time of human plasma. CONCLUSION Overall, this work puts forward inhibitor 1 as a potent and selective inhibitor of FXIIa for further development as an anticoagulant.
Collapse
Affiliation(s)
- Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Daniel K. Afosah
- Department of Chemistry and Biochemistry, Washington and Lee University, Lexington VA 24450, USA
| | | |
Collapse
|
22
|
Yakovleva EV, Zozulya NI. Physiological and pathological role of factor XII. RUSSIAN JOURNAL OF HEMATOLOGY AND TRANSFUSIOLOGY 2022. [DOI: 10.35754/0234-5730-2022-67-4-570-578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction. The most widely accepted notion of the function of blood clotting factor XII (FXII, Hageman factor) is its involvement in the internal blood clotting pathway. However, the biological role of FXII is diverse.Aim – to review the diverse biological functions of FXII.Main findings. FXII is a serine protease. The structure of FXII has a high degree of homology with plasminogen, tissue plasminogen activator and urokinase plasminogen activator. Activated FXII (FXIIa) has five substrates: high-molecular kininogen, precallikrein, FXI, plasminogen, complement proteins (C1s, C1r). FXII provides hemostatic balance by participating in the processes of blood clotting and fibrinolysis. FXII regulates inflammatory and allergic reactions by interacting with the kallikreinkinin system and the complement system. FXII has biological activity in various cells in vivo: endotheliocytes, platelets, monocytes, neutrophils, fi broblasts, dendritic cells, which determines its diverse role in physiological and pathological processes.
Collapse
|
23
|
Imberg L, Platte S, Erbacher C, Daniliuc CG, Kalinina SA, Dörner W, Poso A, Karst U, Kalinin DV. Amide-functionalized 1,2,4-Triazol-5-amines as Covalent Inhibitors of Blood Coagulation Factor XIIa and Thrombin. ACS Pharmacol Transl Sci 2022; 5:1318-1347. [PMID: 36524012 PMCID: PMC9745896 DOI: 10.1021/acsptsci.2c00204] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 12/05/2022]
Abstract
To counteract thrombosis, new safe and efficient antithrombotics are required. We herein report the design, synthesis, and biological activity of a series of amide-functionalized acylated 1,2,4-triazol-5-amines as selective inhibitors of blood coagulation factor XIIa and thrombin. The introduction of an amide moiety into the main scaffold of 3-aryl aminotriazoles added certain three-dimensional properties to synthesized compounds and allowed them to reach binding sites in FXIIa and thrombin previously unaddressed by non-functionalized 1,2,4-triazol-5-amines. Among synthesized compounds, one quinoxaline-derived aminotriazole bearing N-butylamide moiety inhibited FXIIa with the IC50 value of 28 nM, whereas the N-phenylamide-derived aminotriazole inhibited thrombin with the IC50 value of 41 nM. Performed mass-shift experiments and molecular modeling studies proved the covalent mechanism of FXIIa and thrombin inhibition by synthesized compounds. In plasma coagulation tests, developed aminotriazoles showed anticoagulant properties mainly affecting the intrinsic blood coagulation pathway, activation of which is associated with thrombosis but is negligible for hemostasis.
Collapse
Affiliation(s)
- Lukas Imberg
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster 48149, Germany
| | - Simon Platte
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster 48149, Germany
| | - Catharina Erbacher
- Institute
of Inorganic and Analytical Chemistry, University
of Münster, Münster 48149, Germany
| | | | | | - Wolfgang Dörner
- Institute
of Biochemistry, University of Münster, Münster 48149, Germany
| | - Antti Poso
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Department
of Internal Medicine VIII, University Hospital
Tübingen, Tübingen 72076, Germany
| | - Uwe Karst
- Institute
of Inorganic and Analytical Chemistry, University
of Münster, Münster 48149, Germany
| | - Dmitrii V. Kalinin
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster 48149, Germany
| |
Collapse
|
24
|
Dysregulated haemostasis in thrombo-inflammatory disease. Clin Sci (Lond) 2022; 136:1809-1829. [PMID: 36524413 PMCID: PMC9760580 DOI: 10.1042/cs20220208] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Inflammatory disease is often associated with an increased incidence of venous thromboembolism in affected patients, although in most instances, the mechanistic basis for this increased thrombogenicity remains poorly understood. Acute infection, as exemplified by sepsis, malaria and most recently, COVID-19, drives 'immunothrombosis', where the immune defence response to capture and neutralise invading pathogens causes concurrent activation of deleterious prothrombotic cellular and biological responses. Moreover, dysregulated innate and adaptive immune responses in patients with chronic inflammatory conditions, such as inflammatory bowel disease, allergies, and neurodegenerative disorders, are now recognised to occur in parallel with activation of coagulation. In this review, we describe the detailed cellular and biochemical mechanisms that cause inflammation-driven haemostatic dysregulation, including aberrant contact pathway activation, increased tissue factor activity and release, innate immune cell activation and programmed cell death, and T cell-mediated changes in thrombus resolution. In addition, we consider how lifestyle changes increasingly associated with modern life, such as circadian rhythm disruption, chronic stress and old age, are increasingly implicated in unbalancing haemostasis. Finally, we describe the emergence of potential therapies with broad-ranging immunothrombotic functions, and how drug development in this area is challenged by our nascent understanding of the key molecular and cellular parameters that control the shared nodes of proinflammatory and procoagulant pathways. Despite the increasing recognition and understanding of the prothrombotic nature of inflammatory disease, significant challenges remain in effectively managing affected patients, and new therapeutic approaches to curtail the key pathogenic steps in immune response-driven thrombosis are urgently required.
Collapse
|
25
|
Schroeter CB, Rolfes L, Gothan KSS, Gruchot J, Herrmann AM, Bock S, Fazio L, Henes A, Narayanan V, Pfeuffer S, Nelke C, Räuber S, Huntemann N, Duarte-Silva E, Dobelmann V, Hundehege P, Wiendl H, Raba K, Küry P, Kremer D, Ruck T, Müntefering T, Budde T, Cerina M, Meuth SG. Cladribine treatment improves cortical network functionality in a mouse model of autoimmune encephalomyelitis. J Neuroinflammation 2022; 19:270. [DOI: 10.1186/s12974-022-02588-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cladribine is a synthetic purine analogue that interferes with DNA synthesis and repair next to disrupting cellular proliferation in actively dividing lymphocytes. The compound is approved for the treatment of multiple sclerosis (MS). Cladribine can cross the blood–brain barrier, suggesting a potential effect on central nervous system (CNS) resident cells. Here, we explored compartment-specific immunosuppressive as well as potential direct neuroprotective effects of oral cladribine treatment in experimental autoimmune encephalomyelitis (EAE) mice.
Methods
In the current study, we compare immune cell frequencies and phenotypes in the periphery and CNS of EAE mice with distinct grey and white matter lesions (combined active and focal EAE) either orally treated with cladribine or vehicle, using flow cytometry. To evaluate potential direct neuroprotective effects, we assessed the integrity of the primary auditory cortex neuronal network by studying neuronal activity and spontaneous synaptic activity with electrophysiological techniques ex vivo.
Results
Oral cladribine treatment significantly attenuated clinical deficits in EAE mice. Ex vivo flow cytometry showed that cladribine administration led to peripheral immune cell depletion in a compartment-specific manner and reduced immune cell infiltration into the CNS. Histological evaluations revealed no significant differences for inflammatory lesion load following cladribine treatment compared to vehicle control. Single cell electrophysiology in acute brain slices was performed and showed an impact of cladribine treatment on intrinsic cellular firing patterns and spontaneous synaptic transmission in neurons of the primary auditory cortex. Here, cladribine administration in vivo partially restored cortical neuronal network function, reducing action potential firing. Both, the effect on immune cells and neuronal activity were transient.
Conclusions
Our results indicate that cladribine exerts a neuroprotective effect after crossing the blood–brain barrier independently of its peripheral immunosuppressant action.
Collapse
|
26
|
Singh PK, Chen Z, Horn K, Norris EH. Blocking domain 6 of high molecular weight kininogen to understand intrinsic clotting mechanisms. Res Pract Thromb Haemost 2022; 6:e12815. [PMID: 36254255 PMCID: PMC9561425 DOI: 10.1002/rth2.12815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/05/2022] Open
Abstract
Background The contact system is initiated by factor (F) XII activation and the assembly of high molecular weight kininogen (HK) with either FXI or prekallikrein (PK) on a negatively charged surface. Overactivation of this system contributes to thrombosis and inflammation in numerous diseases. To develop effective therapeutics for contact system disorders, a detailed understanding of this pathway is needed. Methods We performed coagulation assays in normal human plasma and various factor-deficient plasmas. To evaluate how HK-mediated PK and FXI activation contributes to coagulation, we used an anti-HK antibody to block access to domain 6 of HK, the region required for efficient activation of PK and FXI. Results FXI's binding to HK and its subsequent activation by activated FXII contributes to coagulation. We found that the 3E8 anti-HK antibody can inhibit the binding of FXI or PK to HK, delaying clot formation in human plasma. Our data show that in the absence of FXI, however, PK can substitute for FXI in this process. Addition of activated FXI (FXIa) or activated PK (PKa) abolished the inhibitory effect of 3E8. Moreover, the requirement of HK in intrinsic coagulation can be largely bypassed by adding FXIa. Like FXIa, exogenous PKa shortened the clotting time in HK-deficient plasma, which was not due to feedback activation of FXII. Conclusions This study improves our understanding of HK-mediated coagulation and provides an explanation for the absence of bleeding in HK-deficient individuals. 3E8 specifically prevented HK-mediated FXI activation; therefore, it could be used to prevent contact activation-mediated thrombosis without altering hemostasis.
Collapse
Affiliation(s)
- Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Zu‐Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Katharina Horn
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
27
|
Bierhansl L, Hartung HP, Aktas O, Ruck T, Roden M, Meuth SG. Thinking outside the box: non-canonical targets in multiple sclerosis. Nat Rev Drug Discov 2022; 21:578-600. [PMID: 35668103 PMCID: PMC9169033 DOI: 10.1038/s41573-022-00477-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system that causes demyelination, axonal degeneration and astrogliosis, resulting in progressive neurological disability. Fuelled by an evolving understanding of MS immunopathogenesis, the range of available immunotherapies for clinical use has expanded over the past two decades. However, MS remains an incurable disease and even targeted immunotherapies often fail to control insidious disease progression, indicating the need for new and exceptional therapeutic options beyond the established immunological landscape. In this Review, we highlight such non-canonical targets in preclinical MS research with a focus on five highly promising areas: oligodendrocytes; the blood-brain barrier; metabolites and cellular metabolism; the coagulation system; and tolerance induction. Recent findings in these areas may guide the field towards novel targets for future therapeutic approaches in MS.
Collapse
Affiliation(s)
- Laura Bierhansl
- Department of Neurology, Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- German Center of Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
28
|
Radoslavova S, Fels B, Pethö Z, Gruner M, Ruck T, Meuth SG, Folcher A, Prevarskaya N, Schwab A, Ouadid-Ahidouch H. TRPC1 channels regulate the activation of pancreatic stellate cells through ERK1/2 and SMAD2 pathways and perpetuate their pressure-mediated activation. Cell Calcium 2022; 106:102621. [PMID: 35905654 DOI: 10.1016/j.ceca.2022.102621] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/02/2022]
Abstract
Pancreatic stellate cell (PSC) activation is a major event occurring during pancreatic ductal adenocarcinoma (PDAC) development. Up to now mechanisms underlying their activation by mechanical cues such as the elevated tissue pressure in PDAC remain poorly understood. Here we investigate the role of one potential mechano-transducer, TRPC1 ion channel, in PSC activation. Using pre-activated human siTRPC1 and murine TRPC1-KO PSCs, we show that TRPC1 promotes αSMA (α-smooth muscle actin) expression, the main activation marker, in cooperation with the phosphorylated SMAD2, under normal and elevated pressure. Functional studies following TRPC1 silencing demonstrate the dual role of TRPC1 in the modulation of PSC proliferation and IL-6 secretion through the activation of ERK1/2 and SMAD2 pathways. Moreover, pressurization changes the mechanical behavior of PSCs by increasing their cellular stiffness and emitted traction forces in a TRPC1-dependent manner. In summary, these results point to a role of TRPC1 channels in sensing and transducing the characteristic mechanical properties of the PDAC microenvironment in PSCs.
Collapse
Affiliation(s)
- Silviya Radoslavova
- Laboratory of Cellular and Molecular Physiology, UR-UPJV 4667, University of Picardie Jules Verne, 80039 Amiens, France; University of Lille, Inserm U1003 - PHYCEL - Cellular Physiology, F-59000 Lille, France
| | - Benedikt Fels
- Institute of Physiology, University Lübeck, Lübeck, Germany; DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Zoltan Pethö
- Institute of Physiology II, University Münster, Münster, Germany
| | - Matthias Gruner
- Institute of Physiology II, University Münster, Münster, Germany
| | - Tobias Ruck
- Klinik für Neurologie, Medical Faculty, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Sven G Meuth
- Klinik für Neurologie, Medical Faculty, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Antoine Folcher
- University of Lille, Inserm U1003 - PHYCEL - Cellular Physiology, F-59000 Lille, France
| | - Natalia Prevarskaya
- University of Lille, Inserm U1003 - PHYCEL - Cellular Physiology, F-59000 Lille, France.
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, Münster, Germany.
| | - Halima Ouadid-Ahidouch
- Laboratory of Cellular and Molecular Physiology, UR-UPJV 4667, University of Picardie Jules Verne, 80039 Amiens, France.
| |
Collapse
|
29
|
Guse K, Hagemann N, Thiele L, Remlinger J, Salmen A, Hoepner R, Keller I, Meyer P, Grandgirard D, Leib SL, Vassella E, Locatelli G, Hermann DM, Chan A. CNS Antigen-Specific Neuroinflammation Attenuates Ischemic Stroke With Involvement of Polarized Myeloid Cells. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2022; 9:9/4/e1168. [PMID: 35676093 PMCID: PMC9177141 DOI: 10.1212/nxi.0000000000001168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 02/25/2022] [Indexed: 11/15/2022]
Abstract
Background and Objectives Experimental studies indicate shared molecular pathomechanisms in cerebral hypoxia-ischemia and autoimmune neuroinflammation. This has led to clinical studies investigating the effects of immunomodulatory therapies approved in multiple sclerosis on inflammatory damage in stroke. So far, mutual and combined interactions of autoimmune, CNS antigen-specific inflammatory reactions and cerebral ischemia have not been investigated so far. Methods Active MOG35-55 experimental autoimmune encephalomyelitis (EAE) was induced in male C57Bl/6J mice. During different phases of EAE, transient middle cerebral artery occlusion (tMCAO, 60 minutes) was induced. Brain tissue was analyzed for infarct size and immune cell infiltration. Multiplex gene expression analysis was performed for 186 genes associated with neuroinflammation and hypoxic-ischemic damage. Results Mice with severe EAE disease showed a substantial reduction in infarct size after tMCAO. Histopathologic analysis showed less infiltration of CD45+ hematopoietic cells in the infarct core of severely diseased acute EAE mice; this was accompanied by an accumulation of Arginase1-positive/Iba1-positive cells. Gene expression analysis indicated an involvement of myeloid cell-driven anti-inflammatory mechanisms in the attenuation of ischemic injury in severely diseased mice exposed to tMCAO in the acute EAE phase. Discussion CNS autoantigen-specific autoimmunity has a protective influence on primary tissue damage after experimental stroke, indicating a very early involvement of CNS antigen-specific, myeloid cell-associated anti-inflammatory immune mechanisms that mitigate ischemic injury in the acute EAE phase.
Collapse
|
30
|
Schaller-Paule MA, Yalachkov Y, Steinmetz H, Friedauer L, Hattingen E, Miesbach W, Weber F, Kirchmayr K, Schaefer JH, Foerch C. Analysis of CSF D-Dimer to Identify Intrathecal Fibrin-Driven Autoimmunity in Patients With Multiple Sclerosis. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2022; 9:9/3/e1150. [PMID: 35260469 PMCID: PMC8906189 DOI: 10.1212/nxi.0000000000001150] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/06/2022] [Indexed: 12/02/2022]
Abstract
Background and Objectives Proteins of the coagulation system contribute to autoimmune inflammation in patients with multiple sclerosis (MS). On blood-brain barrier (BBB) disruption, fibrinogen enters the CNS and is rapidly converted to fibrin, unfolding pleiotropic autoimmune mechanisms. Fibrin accumulation leads to subsequent proteolytic degradation that results in D-dimer generation. The primary objective of this study was to determine intrathecal levels of D-dimer in CSF as a measure of intrathecal coagulation cascade activation and to evaluate its diagnostic utility in patients with MS in contrast to healthy subjects. Key secondary objectives included analysis of CSF D-dimer in differential diagnoses of MS and its relation to routine clinical markers of disease activity. Methods Patients admitted for the assessment of suspected MS were prospectively recruited from October 2017 to December 2020. Blood plasma and citrated CSF samples were analyzed using a highly sensitive luminescent oxygen channeling immunoassay. Intrathecal generation of D-dimer was analyzed by adjusting for CSF/serum albumin (Qalb) and CSF/plasma D-dimer quotients (QD-dimer), and corresponding CSF fibrinogen levels were determined. Final diagnoses after full evaluation and clinical data were recorded. Results Of 187 patients, 113 patients received a diagnosis of MS or clinically/radiologically isolated syndrome. We found increased intrathecal CSF D-dimer generation levels (QD-dimer/Qalb-index) for patients with relapsing-remitting MS (RRMS; n = 71, median 4.7, interquartile range [IQR] 2.5–8.0) when compared with those for disease controls (n = 22, median 2.6, IQR 2.1–4.8, p = 0.031). Absolute CSF D-dimer values correlated with CSF fibrinogen levels (r = 0.463; p < 0 .001) and CSF leukocytes (r = 0.273; p = 0.003) and were elevated in MS patients with contrast enhancement (CE) compared with MS patients without CE on MRI (n = 48, median 6 ng/mL, and IQR 3–15.25 vs n = 41, median 4 ng/mL, and IQR 2–7; p = 0.026). Exploratory subgroup analyses indicated a correlation of intrathecal inflammatory activity and CSF D-dimer levels. Discussion D-dimer in CSF can be reliably determined and correlates with markers of CNS inflammation and CSF fibrinogen levels. Adjusted for BBB dysfunction, CSF D-dimer may allow the identification of intrathecal coagulation cascade activation in patients with MS. Classification of Evidence This study provides Class I evidence that CSF D-dimer levels are elevated in patients with RRMS.
Collapse
|
31
|
F12 as a reliable diagnostic and prognostic biomarker associated with immune infiltration in papillary thyroid cancer. Aging (Albany NY) 2022; 14:3687-3704. [PMID: 35483340 PMCID: PMC9085229 DOI: 10.18632/aging.204037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/14/2022] [Indexed: 11/25/2022]
Abstract
Objective: To explore the function of coagulation factor XII (F12) in papillary thyroid cancer (PTC). Materials and Methods: We assessed F12 expression and its relationship with overall survival (OS) in various cancers using TIMER and TISIDB databases. Further, we evaluated the mRNA and protein expression levels of F12 in PTC via different bioinformatics tools. The receiver operating characteristic (ROC) curve was applied to determine the diagnostic value of F12 in PTC. Then, the Kaplan-Meier plotter and Cox regression analyses were performed to examine the prognostic significance of F12. The possible mechanism of F12 in PTC was investigated through enrichment analyses. Finally, the correlation between F12 expression and immune cell infiltration was analyzed using TCGA data. Results: This study revealed the clinical significance of F12 in various cancers. Higher mRNA (P <0.001) and protein expressions of F12 were observed in PTC compared with normal tissues. Besides, F12 expression exhibited high diagnostic performance in PTC and its overexpression served as an independent predictor for the poor OS (P <0.05). Enrichment analyses results showed that F12 was mainly involved in metabolism-associated pathways. Additionally, F12 expression was significantly linked to immune cell infiltration levels, especially macrophage infiltration. Conclusions: F12 might be a reliable diagnostic and prognostic biomarker for PTC. Moreover, F12 expression might affect the OS of PTC patients via regulating metabolic pathways.
Collapse
|
32
|
Gao H, Liu Y, Hu Y, Ge M, Ding J, Ye Q. Establishment and Application of a Prognostic Risk Score Model Based on Characteristics of Different Immunophenotypes for Lung Adenocarcinoma. Front Genet 2022; 13:850101. [PMID: 35547263 PMCID: PMC9081571 DOI: 10.3389/fgene.2022.850101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/17/2022] [Indexed: 12/24/2022] Open
Abstract
Objective: Lung adenocarcinoma (LUAD) is a highly heterogeneous tumor. Tumor mutations and the immune microenvironment play important roles in LUAD development and progression. This study was aimed at elucidating the characteristics of patients with different tumor immune microenvironment and establishing a prediction model of prognoses and immunotherapy benefits for patients with LUAD. Materials and Methods: We conducted a bioinformatics analysis on data from The Cancer Genome Atlas and Gene Expression Omnibus (training and test sets, respectively). Patients in the training set were clustered into different immunophenotypes based on tumor-infiltrating immune cells (TIICs). The immunophenotypic differentially expressed genes (IDEGs) were used to develop a prognostic risk score (PRS) model. Then, the model was validated in the test set and applied to evaluate 42 surgery patients with early LUAD. Results: Patients in the training set were clustered into high (Immunity_H), medium (Immunity_M), and low (Immunity_L) immunophenotype groups. Immunity_H patients had the best survival and more TIICs than Immunity_L patients. Immunity_M patients had the worst survival, characterized by most CD8+ T and Treg cells and highest expression of PD-1 and PD-L1. The PRS model, which consisted of 14 IDEGs, showed good potential for predicting the prognoses of patients in both training and test sets. In the training set, the low-risk patients had more TIICs, higher immunophenoscores (IPSs) and lower mutation rates of driver genes. The high-risk patients had more mutations of DNA mismatch repair deficiency and APOBEC (apolipoprotein B mRNA editing enzyme catalytic polypeptide-like). The model was also a good indicator of the curative effect for immunotherapy-treated patients. Furthermore, the low-risk group out of 42 patients, which was evaluated by the PRS model, had more TIICs, higher IPSs and better progression-free survival. Additionally, IPSs and PRSs of these patients were correlated with EGFR mutations. Conclusion: The PRS model has good potential for predicting the prognoses and immunotherapy benefits of LUAD patients. It may facilitate the diagnosis, risk stratification, and treatment decision-making for LUAD patients.
Collapse
Affiliation(s)
- Hong Gao
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yanhong Liu
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yue Hu
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Meiling Ge
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jie Ding
- Biobank of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Ye
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
33
|
Abstract
Blood coagulation is essential to maintain the integrity of a closed circulatory system (hemostasis), but also contributes to thromboembolic occlusion of vessels (thrombosis). Thrombosis may cause deep vein thrombosis, pulmonary embolism, myocardial infarction, peripheral artery disease, and ischemic stroke, collectively the most common causes of death and disability in the developed world. Treatment for the prevention of thromboembolic diseases using anticoagulants such as heparin, coumarins, thrombin inhibitors, or antiplatelet drugs increase the risk of bleeding and are associated with an increase in potentially life-threatening hemorrhage, partially offsetting the benefits of reduced coagulation. Thus, drug development aiming at novel targets is needed to provide efficient and safe anticoagulation. Within the last decade, experimental and preclinical data have shown that some coagulation mechanisms principally differ in thrombosis and hemostasis. The plasma contact system protein factors XII and XI, high-molecular-weight kininogen, and plasma kallikrein specifically contribute to thrombosis, however, have minor, if any, role in hemostatic coagulation mechanisms. Inherited deficiency in contact system proteins is not associated with increased bleeding in humans and animal models. Therefore, targeting contact system proteins provides the exciting opportunity to interfere specifically with thromboembolic diseases without increasing the bleeding risk. Recent studies that investigated pharmacologic inhibition of contact system proteins have shown that this approach provides efficient and safe thrombo-protection that in contrast to classical anticoagulants is not associated with increased bleeding risk. This review summarizes therapeutic and conceptual developments for selective interference with pathological thrombus formation, while sparing physiologic hemostasis, that enables safe anticoagulation treatment.
Collapse
Affiliation(s)
- Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Piotr Kuta
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
34
|
Xue C, Lv H, Li Y, Dong N, Wang Y, Zhou J, Shi B, Shan A. Oleanolic acid reshapes the gut microbiota and alters immune-related gene expression of intestinal epithelial cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:764-773. [PMID: 34227118 DOI: 10.1002/jsfa.11410] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/19/2021] [Accepted: 07/05/2021] [Indexed: 05/13/2023]
Abstract
BACKGROUND Oleanolic acid (OA) is a pentacyclic triterpenoid compound that is present at high levels in olive oil and has several promising pharmacological effects, such as liver protection and anti-inflammatory, antioxidant, and anticancer effects. The purpose of the present study was to assess whether OA treatment affects gut health compared to a control condition, including gut microbiota and intestinal epithelial immunity. RESULTS Illumina MiSeq sequencing (16S rRNA gene) was used to investigate the effect of OA on the microbial community of the intestinal tract, while Illumina HiSeq (RNA-seq) technology was used to investigate the regulatory effect of OA on gene expression in intestinal epithelial cells, which allowed for a comprehensive analysis of the effects of OA on intestinal health. The results showed that the consumption of OA initially controlled weight gain in mice and altered the composition of the gut microbiota. At the phylum level, OA significantly increased the relative abundances of cecum Firmicutes but decreased the abundance of Actinobacteria, and at the genus level it increased the relative abundance of potentially beneficial bacteria such as Oscillibacter and Ruminiclostridium 9. Oleanolic acid treatment also altered the expression of 12 genes involved in the Kyoto Encyclopedia of Genes and Genomes(KEGG)pathways of complement and coagulation cascades, hematopoietic cell lineage, and leukocyte transendothelial migration in intestinal epithelial cells to improve gut immunity. CONCLUSION Intake of OA can contribute beneficial effects by optimizing gut microbiota and altering the immune function of intestinal epithelial cells, potentially to improve intestinal health status. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chenyu Xue
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Hao Lv
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Ying Li
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Na Dong
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Yanhui Wang
- The Institute of Animal Nutrition, Heilongjiang Polytechnic, Shuangcheng, P. R. China
| | - Jiale Zhou
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Baoming Shi
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| | - Anshan Shan
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, P. R. China
| |
Collapse
|
35
|
Shavit-Stein E, Berkowitz S, Gofrit SG, Altman K, Weinberg N, Maggio N. Neurocoagulation from a Mechanistic Point of View in the Central Nervous System. Semin Thromb Hemost 2022; 48:277-287. [PMID: 35052009 DOI: 10.1055/s-0041-1741569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coagulation mechanisms are critical for maintaining homeostasis in the central nervous system (CNS). Thrombin, an important player of the coagulation cascade, activates protease activator receptors (PARs), members of the G-protein coupled receptor family. PAR1 is located on neurons and glia. Following thrombin activation, PAR1 signals through the extracellular signal-regulated kinase pathway, causing alterations in neuronal glutamate release and astrocytic morphological changes. Similarly, the anticoagulation factor activated protein C (aPC) can cleave PAR1, following interaction with the endothelial protein C receptor. Both thrombin and aPC are expressed on endothelial cells and pericytes in the blood-brain barrier (BBB). Thrombin-induced PAR1 activation increases cytosolic Ca2+ concentration in brain vessels, resulting in nitric oxide release and increasing F-actin stress fibers, damaging BBB integrity. aPC also induces PAR1 activation and preserves BBB vascular integrity via coupling to sphingosine 1 phosphate receptors. Thrombin-induced PAR1 overactivation and BBB disruption are evident in CNS pathologies. During epileptic seizures, BBB disruption promotes thrombin penetration. Thrombin induces PAR1 activation and potentiates N-methyl-D-aspartate receptors, inducing glutamate-mediated hyperexcitability. Specific PAR1 inhibition decreases status epilepticus severity in vivo. In stroke, the elevation of brain thrombin levels further compromises BBB integrity, with direct parenchymal damage, while systemic factor Xa inhibition improves neurological outcomes. In multiple sclerosis (MS), brain thrombin inhibitory capacity correlates with clinical presentation. Both thrombin inhibition by hirudin and the use of recombinant aPC improve disease severity in an MS animal model. This review presents the mechanisms underlying the effects of coagulation on the physiology and pathophysiology of the CNS.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Keren Altman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nitai Weinberg
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
36
|
Kim H, Shin Y, Kim DH. Mechanobiological Implications of Cancer Progression in Space. Front Cell Dev Biol 2021; 9:740009. [PMID: 34957091 PMCID: PMC8692837 DOI: 10.3389/fcell.2021.740009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
The human body is normally adapted to maintain homeostasis in a terrestrial environment. The novel conditions of a space environment introduce challenges that changes the cellular response to its surroundings. Such an alteration causes physical changes in the extracellular microenvironment, inducing the secretion of cytokines such as interleukin-6 (IL-6) and tumor growth factor-β (TGF-β) from cancer cells to enhance cancer malignancy. Cancer is one of the most prominent cell types to be affected by mechanical cues via active interaction with the tumor microenvironment. However, the mechanism by which cancer cells mechanotransduce in the space environment, as well as the influence of this process on human health, have not been fully elucidated. Due to the growing interest in space biology, this article reviews cancer cell responses to the representative conditions altered in space: microgravity, decompression, and irradiation. Interestingly, cytokine and gene expression that assist in tumor survival, invasive phenotypic transformation, and cancer cell proliferation are upregulated when exposed to both simulated and actual space conditions. The necessity of further research on space mechanobiology such as simulating more complex in vivo experiments or finding other mechanical cues that may be encountered during spaceflight are emphasized.
Collapse
Affiliation(s)
- Hyondeog Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - Yun Shin
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea.,Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, South Korea
| |
Collapse
|
37
|
Chen J, Zhang R, Xie M, Luan C, Li X. Transcriptome Sequencing Identifies PLAUR as an Important Player in Patients With Dermatomyositis-Associated Interstitial Lung Disease. Front Genet 2021; 12:784215. [PMID: 34938325 PMCID: PMC8685457 DOI: 10.3389/fgene.2021.784215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/03/2021] [Indexed: 11/15/2022] Open
Abstract
Dermatomyositis (DM), an inflammatory disorder, is often associated with interstitial lung disease (ILD). However, the underlying mechanism remains unclear. Our study performed RNA sequencing (RNA-seq) and integrative bioinformatics analysis of differentially expressed genes (DEGs) in patients with dermatomyositis-associated interstitial lung disease (DM-ILD) and healthy controls. A total of 2,018 DEGs were identified between DM-ILD and healthy blood samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that DEGs were mainly involved in immune- and inflammatory-related biological processes and pathways. Disease ontology (DO) enrichment analysis identified 35 candidate key genes involved in both skin and lung diseases. Meanwhile, a total of 886 differentially expressed alternative splicing (AS) events were found between DM-ILD and healthy blood samples. After overlapping DEGs with differential AS genes, the plasminogen activator and urokinase receptor (PLAUR) involved in immune-related biological processes and complement and coagulation cascades was screened and identified as the most important gene associated with DM-ILD. The protein–protein interaction (PPI) network revealed that PLAUR had interactions with multiple candidate key genes. Moreover, we observed that there were significantly more neutrophils and less naive B cells in DM-ILD samples than in healthy samples. And the expression of PLAUR was significantly positively correlated with the abundance of neutrophils. Significant higher abundance of PLAUR in DM-ILD patients than healthy controls was validated by RT-qPCR. In conclusion, we identified PLAUR as an important player in regulating DM-ILD by neutrophil-associated immune response. These findings enrich our understanding, which may benefit DM-ILD patients.
Collapse
Affiliation(s)
- Juan Chen
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruixian Zhang
- The Center for Disease Control and Prevention of Yunnan Province, Kunming, China
| | - Min Xie
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chunyan Luan
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaolan Li
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
38
|
Mailer RK, Rangaswamy C, Konrath S, Emsley J, Renné T. An update on factor XII-driven vascular inflammation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119166. [PMID: 34699874 DOI: 10.1016/j.bbamcr.2021.119166] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022]
Abstract
The plasma protein factor XII (FXII) is the liver-derived zymogen of the serine protease FXIIa that initiates an array of proteolytic cascades. Zymogen activation, enzymatic FXIIa activity and functions are regulated by interactions with cell receptors, negatively charged surfaces, other serine proteases, and serpin inhibitors, which bind to distinct protein domains and regions in FXII(a). FXII exerts mitogenic activity, while FXIIa initiates the pro-inflammatory kallikrein-kinin pathway and the pro-thrombotic intrinsic coagulation pathway, respectively. Growing evidence indicates that FXIIa-mediated thrombo-inflammation plays a crucial role in various pathological states besides classical thrombosis, such as endothelial dysfunction. Consistently, increased FXIIa levels are associated with hypercholesterolemia and hypertriglyceridemia. In contrast, FXII deficiency protects from thrombosis but is otherwise not associated with prolonged bleeding or other adverse clinical manifestations. Here, we review current concepts for FXII(a)-driven vascular inflammation focusing on endothelial hyperpermeability, receptor signaling, atherosclerosis and immune cell activation.
Collapse
Affiliation(s)
- Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Emsley
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany.
| |
Collapse
|
39
|
Schroeter CB, Huntemann N, Bock S, Nelke C, Kremer D, Pfeffer K, Meuth SG, Ruck T. Crosstalk of Microorganisms and Immune Responses in Autoimmune Neuroinflammation: A Focus on Regulatory T Cells. Front Immunol 2021; 12:747143. [PMID: 34691057 PMCID: PMC8529161 DOI: 10.3389/fimmu.2021.747143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are the major determinant of peripheral immune tolerance. Many Treg subsets have been described, however thymus-derived and peripherally induced Tregs remain the most important subpopulations. In multiple sclerosis, a prototypical autoimmune disorder of the central nervous system, Treg dysfunction is a pathogenic hallmark. In contrast, induction of Treg proliferation and enhancement of their function are central immune evasion mechanisms of infectious pathogens. In accordance, Treg expansion is compartmentalized to tissues with high viral replication and prolonged in chronic infections. In friend retrovirus infection, Treg expansion is mainly based on excessive interleukin-2 production by infected effector T cells. Moreover, pathogens seem also to enhance Treg functions as shown in human immunodeficiency virus infection, where Tregs express higher levels of effector molecules such as cytotoxic T-lymphocyte-associated protein 4, CD39 and cAMP and show increased suppressive capacity. Thus, insights into the molecular mechanisms by which intracellular pathogens alter Treg functions might aid to find new therapeutic approaches to target central nervous system autoimmunity. In this review, we summarize the current knowledge of the role of pathogens for Treg function in the context of autoimmune neuroinflammation. We discuss the mechanistic implications for future therapies and provide an outlook for new research directions.
Collapse
Affiliation(s)
- Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefanie Bock
- Department of Neurology With Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
40
|
Searle AK, Chen YC, Wallert M, McFadyen J, Maluenda A, Noonan J, Kanellakis P, Zaldivia MT, Huang A, Lioe H, Biondo M, Nolte MW, Rossato P, Bobik A, Panousis C, Wang X, Hosseini H, Peter K. Pharmacological inhibition of Factor XIIa attenuates abdominal aortic aneurysm, reduces atherosclerosis, and stabilizes atherosclerotic plaques. Thromb Haemost 2021; 122:196-207. [PMID: 34619795 PMCID: PMC8820844 DOI: 10.1055/a-1663-8208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background
3F7 is a monoclonal antibody targeting the enzymatic pocket of activated factor XII (FXIIa), thereby inhibiting its catalytic activity. Given the emerging role of FXIIa in promoting thromboinflammation, along with its apparent redundancy for hemostasis, the selective inhibition of FXIIa represents a novel and highly attractive approach targeting pathogenic processes that cause thromboinflammation-driven cardiovascular diseases.
Methods
The effects of FXIIa inhibition were investigated using three distinct mouse models of cardiovascular disease—angiotensin II-induced abdominal aortic aneurysm (AAA), an ApoE
−/−
model of atherosclerosis, and a tandem stenosis model of atherosclerotic plaque instability. 3F7 or its isotype control, BM4, was administered to mice (10 mg/kg) on alternate days for 4 to 8 weeks, depending on the experimental model. Mice were examined for the development and size of AAAs, or the burden and instability of atherosclerosis and associated markers of inflammation.
Results
Inhibition of FXIIa resulted in a reduced incidence of larger AAAs, with less acute aortic ruptures and an associated fibro-protective phenotype. FXIIa inhibition also decreased stable atherosclerotic plaque burden and achieved plaque stabilization associated with increased deposition of fibrous structures, a >2-fold thicker fibrous cap, increased cap-to-core ratio, and reduction in localized and systemic inflammatory markers.
Conclusion
Inhibition of FXIIa attenuates disease severity across three mouse models of thromboinflammation-driven cardiovascular diseases. Specifically, the FXIIa-inhibiting monoclonal antibody 3F7 reduces AAA severity, inhibits the development of atherosclerosis, and stabilizes vulnerable plaques. Ultimately, clinical trials in patients with cardiovascular diseases such as AAA and atherosclerosis are warranted to demonstrate the therapeutic potential of FXIIa inhibition.
Collapse
Affiliation(s)
- Amy K Searle
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Yung Chih Chen
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Maria Wallert
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - James McFadyen
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Ana Maluenda
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Jonathan Noonan
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Peter Kanellakis
- Atherosclerosis and Cell Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Maria Tk Zaldivia
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Angela Huang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Hadi Lioe
- Bio21 Institute, CSL Limited, Parkville, Australia
| | - Mark Biondo
- Bio21 Institute, CSL Limited, Parkville, Australia
| | | | | | - Alex Bobik
- Atherosclerosis and Cell Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Con Panousis
- Bio21 Institute, CSL Limited, Parkville, Australia
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Hamid Hosseini
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart Research Institute - BHRI, Melbourne, Australia
| |
Collapse
|
41
|
Abbadessa G, Lavorgna L, Treaba CA, Bonavita S, Mainero C. Hemostatic factors in the pathogenesis of neuroinflammation in multiple sclerosis. Mult Scler 2021; 28:1834-1842. [PMID: 34410198 DOI: 10.1177/13524585211039111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND A growing body of evidence has shed light on the role of the hemostatic pathway and its components in the pathogenesis of multiple sclerosis (MS), particularly in enhancing and sustaining neuroinflammation. OBJECTIVE To review the clinical, experimental, and neuroimaging evidence supporting the role of different components of the hemostatic pathway in the pathogenesis of neuroinflammation in MS and discuss their translational potential as disease biomarkers and therapeutic targets. METHODS A literature search for most relevant articles from 1956 to 2020 was conducted in PubMed and Scopus. RESULTS Hemostasis components appear to be involved in different key events of neuroinflammation in MS including mononuclear cell diapedesis, microglia activation, and neuronal damage. CONCLUSION The findings on the interplay between hemostatic and thrombotic molecular pathways in the pathogenesis of neuroinflammation in MS open new opportunities for developing novel biomarkers for disease monitoring and prognosis, as well as novel therapeutic targets.
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Luigi Lavorgna
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Constantina Andrada Treaba
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA/Harvard Medical School, MA, USA
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Caterina Mainero
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA/Harvard Medical School, MA, USA
| |
Collapse
|
42
|
Abstract
Introduction: Blood coagulation factor XII (FXII) is an emerging and potentially safe drug target, which dysregulation is associated with thrombosis, hereditary angioedema, and (neuro)inflammation. At the same time, FXII-deficiency is practically asymptomatic. Industrial and academic institutions have developed a number of potential therapeutic agents targeting either FXII zymogen or its active form FXIIa for the treatment of thrombotic and inflammatory conditions associated with the activity of this enzyme.Areas covered: A short overview of the FXII(a) structure and function, underlining its suitability as a drug target, is given. The article reviews patents reported over the last three decades on FXII(a)-targeting therapeutic agents. These agents include small molecules, proteins, peptides, oligonucleotides, siRNAs, and monoclonal antibodies.Expert opinion: The performed analysis of patents revealed that many FXII(a) inhibitors are in the early preclinical stage, while several already showed efficacy in vivo animal models of thrombosis, sepsis, hereditary angioedema, and multiple sclerosis. Two anti-FXIIa agents namely tick protein Ir-CPI and monoclonal antibody CSL312 are currently in human clinical trials. The results of these trials and further studies of FXII(a) pathophysiological functions will encourage the development of new FXII(a) inhibitors.
Collapse
Affiliation(s)
- Dmitrii V Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| |
Collapse
|
43
|
Rangaswamy C, Mailer RK, Englert H, Konrath S, Renné T. The contact system in liver injury. Semin Immunopathol 2021; 43:507-517. [PMID: 34125270 PMCID: PMC8202222 DOI: 10.1007/s00281-021-00876-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/27/2021] [Indexed: 01/18/2023]
Abstract
Coagulation is controlled by a delicate balance of prothrombotic and antithrombotic mechanisms, to prevent both excessive blood loss from injured vessels and pathologic thrombosis. The liver plays a pivotal role in hemostasis through the synthesis of plasma coagulation factors and their inhibitors that, in addition to thrombosis and hemostasis, orchestrates an array of inflammatory responses. As a result, impaired liver function has been linked with both hypercoagulability and bleeding disorders due to a pathologic balance of pro- and anticoagulant plasma factors. At sites of vascular injury, thrombus propagation that finally may occlude the blood vessel depends on negatively charged biopolymers, such as polyphosphates and extracellular DNA, that provide a physiological surface for contact activation of coagulation factor XII (FXII). FXII initiates the contact system that drives both the intrinsic pathway of coagulation, and formation of the inflammatory mediator bradykinin by the kallikrein–kinin system. Moreover, FXII facilitates receptor-mediated signalling, thereby promoting mitogenic activities, angiogenesis, and neutrophil stimulation with implications for liver diseases. Here, we summarize current knowledge on the FXII-driven contact system in liver diseases and review therapeutic approaches to target its activities during impaired liver function.
Collapse
Affiliation(s)
- Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Hanna Englert
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany.
| |
Collapse
|
44
|
Weiland J, Beez A, Westermaier T, Kunze E, Sirén AL, Lilla N. Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH). Int J Mol Sci 2021; 22:5442. [PMID: 34064048 PMCID: PMC8196706 DOI: 10.3390/ijms22115442] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) remains a disease with high mortality and morbidity. Since treating vasospasm has not inevitably led to an improvement in outcome, the actual emphasis is on finding neuroprotective therapies in the early phase following aSAH to prevent secondary brain injury in the later phase of disease. Within the early phase, neuroinflammation, thromboinflammation, disturbances in brain metabolism and early neuroprotective therapies directed against delayed cerebral ischemia (DCI) came into focus. Herein, the role of neuroinflammation, thromboinflammation and metabolism in aSAH is depicted. Potential neuroprotective strategies regarding neuroinflammation target microglia activation, metalloproteases, autophagy and the pathway via Toll-like receptor 4 (TLR4), high mobility group box 1 (HMGB1), NF-κB and finally the release of cytokines like TNFα or IL-1. Following the link to thromboinflammation, potential neuroprotective therapies try to target microthrombus formation, platelets and platelet receptors as well as clot clearance and immune cell infiltration. Potential neuroprotective strategies regarding metabolism try to re-balance the mismatch of energy need and supply following aSAH, for example, in restoring fuel to the TCA cycle or bypassing distinct energy pathways. Overall, this review addresses current neuroprotective strategies in aSAH, hopefully leading to future translational therapy options to prevent secondary brain injury.
Collapse
Affiliation(s)
- Judith Weiland
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Alexandra Beez
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Thomas Westermaier
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
- Department of Neurosurgery, Helios-Amper Klinikum Dachau, Krankenhausstr. 15, 85221 Dachau, Germany
| | - Ekkehard Kunze
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
- Department of Neurosurgery, University Hospital Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
45
|
Stetter C, Lopez-Caperuchipi S, Hopp-Krämer S, Bieber M, Kleinschnitz C, Sirén AL, Albert-Weißenberger C. Amelioration of Cognitive and Behavioral Deficits after Traumatic Brain Injury in Coagulation Factor XII Deficient Mice. Int J Mol Sci 2021; 22:4855. [PMID: 34063730 PMCID: PMC8124758 DOI: 10.3390/ijms22094855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/01/2021] [Indexed: 11/23/2022] Open
Abstract
Based on recent findings that show that depletion of factor XII (FXII) leads to better posttraumatic neurological recovery, we studied the effect of FXII-deficiency on post-traumatic cognitive and behavioral outcomes in female and male mice. In agreement with our previous findings, neurological deficits on day 7 after weight-drop traumatic brain injury (TBI) were significantly reduced in FXII-/- mice compared to wild type (WT) mice. Also, glycoprotein Ib (GPIb)-positive platelet aggregates were more frequent in brain microvasculature of WT than FXII-/- mice 3 months after TBI. Six weeks after TBI, memory for novel object was significantly reduced in both female and male WT but not in FXII-/- mice compared to sham-operated mice. In the setting of automated home-cage monitoring of socially housed mice in IntelliCages, female WT mice but not FXII-/- mice showed decreased exploration and reacted negatively to reward extinction one month after TBI. Since neuroendocrine stress after TBI might contribute to trauma-induced cognitive dysfunction and negative emotional contrast reactions, we measured peripheral corticosterone levels and the ration of heart, lung, and spleen weight to bodyweight. Three months after TBI, plasma corticosterone levels were significantly suppressed in both female and male WT but not in FXII-/- mice, while the relative heart weight increased in males but not in females of both phenotypes when compared to sham-operated mice. Our results indicate that FXII deficiency is associated with efficient post-traumatic behavioral and neuroendocrine recovery.
Collapse
Affiliation(s)
- Christian Stetter
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
| | - Simon Lopez-Caperuchipi
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
| | - Sarah Hopp-Krämer
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (M.B.); (C.K.)
| | - Michael Bieber
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (M.B.); (C.K.)
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (M.B.); (C.K.)
- Department of Neurology and Center for Translational and Behavioral Neurosciences (C-TNBS), University Hospital of Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
| | - Christiane Albert-Weißenberger
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany; (C.S.); (S.L.-C.); (S.H.-K.); (C.A.-W.)
- Institute for Physiology, Department for Neurophysiology, Julius-Maximilians-University Würzburg, Röntgenring 9, 97070 Würzburg, Germany
| |
Collapse
|
46
|
Shlobin NA, Har-Even M, Itsekson-Hayosh Z, Harnof S, Pick CG. Role of Thrombin in Central Nervous System Injury and Disease. Biomolecules 2021; 11:562. [PMID: 33921354 PMCID: PMC8070021 DOI: 10.3390/biom11040562] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Thrombin is a Na+-activated allosteric serine protease of the chymotrypsin family involved in coagulation, inflammation, cell protection, and apoptosis. Increasingly, the role of thrombin in the brain has been explored. Low concentrations of thrombin are neuroprotective, while high concentrations exert pathological effects. However, greater attention regarding the involvement of thrombin in normal and pathological processes in the central nervous system is warranted. In this review, we explore the mechanisms of thrombin action, localization, and functions in the central nervous system and describe the involvement of thrombin in stroke and intracerebral hemorrhage, neurodegenerative diseases, epilepsy, traumatic brain injury, and primary central nervous system tumors. We aim to comprehensively characterize the role of thrombin in neurological disease and injury.
Collapse
Affiliation(s)
- Nathan A. Shlobin
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Meirav Har-Even
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ze’ev Itsekson-Hayosh
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel;
- Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer 5262000, Israel
| | - Sagi Harnof
- Department of Neurosurgery, Beilinson Hospital, Rabin Medical Center, Tel Aviv University, Petah Tikva 4941492, Israel;
| | - Chaim G. Pick
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Center for Biology of Addictive Diseases, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
47
|
Jordan KR, Parra-Izquierdo I, Gruber A, Shatzel JJ, Pham P, Sherman LS, McCarty OJT, Verbout NG. Thrombin generation and activity in multiple sclerosis. Metab Brain Dis 2021; 36:407-420. [PMID: 33411219 PMCID: PMC7864536 DOI: 10.1007/s11011-020-00652-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/25/2020] [Indexed: 01/19/2023]
Abstract
The coagulation cascade and immune system are intricately linked, highly regulated and respond cooperatively in response to injury and infection. Increasingly, evidence of hyper-coagulation has been associated with autoimmune disorders, including multiple sclerosis (MS). The pathophysiology of MS includes immune cell activation and recruitment to the central nervous system (CNS) where they degrade myelin sheaths, leaving neuronal axons exposed to damaging inflammatory mediators. Breakdown of the blood-brain barrier (BBB) facilitates the entry of peripheral immune cells. Evidence of thrombin activity has been identified within the CNS of MS patients and studies using animal models of experimental autoimmune encephalomyelitis (EAE), suggest increased thrombin generation and activity may play a role in the pathogenesis of MS as well as inhibit remyelination processes. Thrombin is a serine protease capable of cleaving multiple substrates, including protease activated receptors (PARs), fibrinogen, and protein C. Cleavage of all three of these substrates represent pathways through which thrombin activity may exert immuno-regulatory effects and regulate permeability of the BBB during MS and EAE. In this review, we summarize evidence that thrombin activity directly, through PARs, and indirectly, through fibrin formation and activation of protein C influences neuro-immune responses associated with MS and EAE pathology.
Collapse
Affiliation(s)
- Kelley R Jordan
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA.
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
| | - András Gruber
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
- Aronora Inc, Portland, OR, USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
| | - Peter Pham
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
| | - Norah G Verbout
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Aronora Inc, Portland, OR, USA
| |
Collapse
|
48
|
Saksida T, Jevtić B, Djedović N, Miljković Đ, Stojanović I. Redox Regulation of Tolerogenic Dendritic Cells and Regulatory T Cells in the Pathogenesis and Therapy of Autoimmunity. Antioxid Redox Signal 2021; 34:364-382. [PMID: 32458699 DOI: 10.1089/ars.2019.7999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Autoimmune diseases are progressively affecting westernized societies, as the proportion of individuals suffering from autoimmunity is steadily increasing over the past decades. Understanding the role of reactive oxygen species (ROS) in modulation of the immune response in the pathogenesis of autoimmune disorders is of utmost importance. The focus of this review is the regulation of ROS production within tolerogenic dendritic cells (tolDCs) and regulatory T (Treg) cells that have the essential role in the prevention of autoimmune diseases and significant potency in their therapy. Recent Advances: It is now clear that ROS are extremely important for the proper function of both DC and T cells. Antigen processing/presentation and the ability of DC to activate T cells depend upon the ROS availability. Treg differentiation, suppressive function, and stability are profoundly influenced by ROS presence. Critical Issues: Although a plethora of results on the relation between ROS and immune cells exist, it remains unclear whether ROS modulation is a productive way for skewing T cells and DCs toward a tolerogenic phenotype. Also, the possibility of ROS modulation for enhancement of regulatory properties of DC and Treg during their preparation for use in cellular therapy has to be clarified. Future Directions: Studies of DC and T cell redox regulation should allow for the improvement of the therapy of autoimmune diseases. This could be achieved through the direct therapeutic application of ROS modulators in autoimmunity, or indirectly through ROS-dependent enhancement of tolDC and Treg preparation for cell-based immunotherapy. Antioxid. Redox Signal. 34, 364-382.
Collapse
Affiliation(s)
- Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Neda Djedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
49
|
Factor XII/XIIa inhibitors: Their discovery, development, and potential indications. Eur J Med Chem 2020; 208:112753. [DOI: 10.1016/j.ejmech.2020.112753] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022]
|
50
|
Oswald JT, Patel H, Khan D, Jeorje NN, Golzar H, Oswald EL, Tang S. Drug Delivery Systems Using Surface Markers for Targeting Cancer Stem Cells. Curr Pharm Des 2020; 26:2057-2071. [PMID: 32250211 DOI: 10.2174/1381612826666200406084900] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
Abstract
The innate abilities of cancer stem cells (CSCs), such as multi-drug resistance, drug efflux, quiescence and ionizing radiation tolerance, protect them from most traditional chemotherapeutics. As a result, this small subpopulation of persistent cells leads to more aggressive and chemoresistant cancers, causing tumour relapse and metastasis. This subpopulation is differentiated from the bulk tumour population through a wide variety of surface markers expressed on the cell surface. Recent developments in nanomedicine and targeting delivery methods have given rise to new possibilities for specifically targeting these markers and preferentially eliminating CSCs. Herein, we first summarize the range of surface markers identifying CSC populations in a variety of cancers; then, we discuss recent attempts to actively target CSCs and their niches using liposomal, nanoparticle, carbon nanotube and viral formulations.
Collapse
Affiliation(s)
- James T Oswald
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Haritosh Patel
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Daid Khan
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ninweh N Jeorje
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Hossein Golzar
- Department of Chemistry & Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Erin L Oswald
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Shirley Tang
- Department of Chemistry & Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|