1
|
Kawakita F, Nakajima H, Suzuki Y, Nampei M, Oinaka H, Suzuki H. Effects of Haptoglobin on Early Brain Injury, Vasospasm, and Lymphatic Drainage After Subarachnoid Hemorrhage in Mice. Stroke 2024; 55:2885-2895. [PMID: 39474674 PMCID: PMC11593995 DOI: 10.1161/strokeaha.124.048048] [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: 06/03/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Erythrolysis releases free Hb (hemoglobin), which is one of the most upstream and important molecules causing early brain injury and cerebral vasospasm after subarachnoid hemorrhage (SAH). The purpose of this study was to investigate if a Hb scavenger protein Hp (haptoglobin) supplementation prevents early brain injury and cerebral vasospasm and influences the lymphatic drainage mechanism in an established SAH model of mice by a blood injection into the prechiasmatic cistern. METHODS This study consisted of 4 parts, with a total of 317 C57BL/6 male mice undergoing sham or SAH modeling, randomly followed by 24-hour intracerebroventricular infusion of no vehicle, 30.1 mg/mL BSA, 30.1 mg/mL Hp (100%; a mixture of Hp1-1, Hp2-1, and Hp2-2), 50% Hp, 25% Hp, and 12.5% Hp solutions from 30 minutes postmodeling. The effects were evaluated at 24 and 48 hours postmodeling. RESULTS The 100%Hp decreased mortality and brain edema until 48 hours post-SAH and suppressed post-SAH neurological impairments, Hb infiltrations into the perivascular spaces and brain tissues, fibrinogen-positive microthrombi formation, microglial activation, and caspase-dependent neuronal apoptosis, as well as large-vessel vasospasm on India-ink angiography at 24 hours compared with vehicle-treated SAH mice. The Hp solutions up to 50% concentrations also prevented post-SAH neurological impairments, and those up to 25% concentrations suppressed post-SAH neuronal apoptosis and vasospasm development until 48 hours. Immunohistochemical staining of deep cervical and mandibular lymph nodes demonstrated that Hb was increased in the lymphatic sinus after SAH and was further increased by Hp administration in SAH animals. CONCLUSIONS This study first showed that an Hp concentrate prevented early brain injury and cerebral vasospasm by inhibiting Hb penetration into brain tissues and increasing lymphatic drainage of free Hb in an established SAH model of mice.
Collapse
Affiliation(s)
- Fumihiro Kawakita
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hideki Nakajima
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yume Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Mai Nampei
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroki Oinaka
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
2
|
Soladogun AS, Zhang L. The Neural Palette of Heme: Altered Heme Homeostasis Underlies Defective Neurotransmission, Increased Oxidative Stress, and Disease Pathogenesis. Antioxidants (Basel) 2024; 13:1441. [PMID: 39765770 PMCID: PMC11672823 DOI: 10.3390/antiox13121441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
Heme, a complex iron-containing molecule, is traditionally recognized for its pivotal role in oxygen transport and cellular respiration. However, emerging research has illuminated its multifaceted functions in the nervous system, extending beyond its canonical roles. This review delves into the diverse roles of heme in the nervous system, highlighting its involvement in neural development, neurotransmission, and neuroprotection. We discuss the molecular mechanisms by which heme modulates neuronal activity and synaptic plasticity, emphasizing its influence on ion channels and neurotransmitter receptors. Additionally, the review explores the potential neuroprotective properties of heme, examining its role in mitigating oxidative stress, including mitochondrial oxidative stress, and its implications in neurodegenerative diseases. Furthermore, we address the pathological consequences of heme dysregulation, linking it to conditions such as Alzheimer's disease, Parkinson's disease, and traumatic brain injuries. By providing a comprehensive overview of heme's multifunctional roles in the nervous system, this review underscores its significance as a potential therapeutic target and diagnostic biomarker for various neurological disorders.
Collapse
Affiliation(s)
| | - Li Zhang
- Department of Biological Sciences, School of Natural Sciences and Mathematics, University of Texas at Dallas, Richardson, TX 75080, USA;
| |
Collapse
|
3
|
Pedard M, Prevost L, Carpena C, Holleran B, Desrues L, Dubois M, Nicola C, Gruel R, Godefroy D, Deffieux T, Tanter M, Ali C, Leduc R, Prézeau L, Gandolfo P, Morin F, Wurtz O, Bonnard T, Vivien D, Castel H. The urotensin II receptor triggers an early meningeal response and a delayed macrophage-dependent vasospasm after subarachnoid hemorrhage in male mice. Nat Commun 2024; 15:8430. [PMID: 39341842 PMCID: PMC11439053 DOI: 10.1038/s41467-024-52654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Subarachnoid hemorrhage (SAH) can be associated with neurological deficits and has profound consequences for mortality and morbidity. Cerebral vasospasm (CVS) and delayed cerebral ischemia affect neurological outcomes in SAH patients, but their mechanisms are not fully understood, and effective treatments are limited. Here, we report that urotensin II receptor UT plays a pivotal role in both early events and delayed mechanisms following SAH in male mice. Few days post-SAH, UT expression is triggered by blood or hemoglobin in the leptomeningeal compartment. UT contributes to perimeningeal glia limitans astrocyte reactivity, microvascular alterations and neuroinflammation independent of CNS-associated macrophages (CAMs). Later, CAM-dependent vascular inflammation and subsequent CVS develop, leading to cognitive dysfunction. In an SAH model using humanized UTh+/h+ male mice, we show that post-SAH CVS and behavioral deficits, mediated by UT through Gq/PLC/Ca2+ signaling, are prevented by UT antagonists. These results highlight the potential of targeting UT pathways to reduce early meningeal response and delayed cerebral ischemia in SAH patients.
Collapse
Affiliation(s)
- Martin Pedard
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Lucie Prevost
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Camille Carpena
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Brian Holleran
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Laurence Desrues
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Martine Dubois
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Celeste Nicola
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Roxane Gruel
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - David Godefroy
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
- Univ Rouen Normandie, Inserm, Normandie Univ, NorDiC UMR 1239, Rouen, France
| | - Thomas Deffieux
- Institute Physics for Medicine, Inserm U1273, CNRS UMR 8631, ESPCI Paris, Paris Sciences et Lettres PSL University, Paris, France
| | - Mickael Tanter
- Institute Physics for Medicine, Inserm U1273, CNRS UMR 8631, ESPCI Paris, Paris Sciences et Lettres PSL University, Paris, France
| | - Carine Ali
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Laurent Prézeau
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Pierrick Gandolfo
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Fabrice Morin
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Olivier Wurtz
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Thomas Bonnard
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
- Centre Hospitalier Universitaire Caen, Department of Clinical Research, Caen, France
| | - Hélène Castel
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France.
| |
Collapse
|
4
|
Morshed N, Rennie C, Deng W, Collins-Praino L, Care A. Serum-derived protein coronas affect nanoparticle interactions with brain cells. NANOTECHNOLOGY 2024; 35:495101. [PMID: 39284320 DOI: 10.1088/1361-6528/ad7b40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/16/2024] [Indexed: 09/20/2024]
Abstract
Neuronanomedicine is an emerging field bridging the gap between neuromedicine and novel nanotherapeutics. Despite promise, clinical translation of neuronanomedicine remains elusive, possibly due to a dearth of information regarding the effect of the protein corona on these neuronanomedicines. The protein corona, a layer of proteins adsorbed to nanoparticles following exposure to biological fluids, ultimately determines the fate of nanoparticles in biological systems, dictating nanoparticle-cell interactions. To date, few studies have investigated the effect of the protein corona on interactions with brain-derived cells, an important consideration for the development of neuronanomedicines. Here, two polymeric nanoparticles, poly(lactic-co-glycolic acid) (PLGA) and PLGA-polyethylene glycol (PLGA-PEG), were used to obtain serum-derived protein coronas. Protein corona characterization and liquid chromatography mass spectrometry analysis revealed distinct differences in biophysical properties and protein composition. PLGA protein coronas contained high abundance of globins (60%) and apolipoproteins (21%), while PLGA-PEG protein coronas contained fewer globins (42%) and high abundance of protease inhibitors (28%). Corona coated PLGA nanoparticles were readily internalized into microglia and neuronal cells, but not into astrocytes. Internalization of nanoparticles was associated with pro-inflammatory cytokine release and decreased neuronal cell viability, however, viability was rescued in cells treated with corona coated nanoparticles. These results showcase the importance of the protein corona in mediating nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Nabila Morshed
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Claire Rennie
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Gadigal Country, Sydney, NSW 2007, Australia
| | - Lyndsey Collins-Praino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
- Biologics Innovation Facility, University of Technology Sydney, Gadigal Country, Sydney, NSW 2007, Australia
| |
Collapse
|
5
|
Gardner RS, Kyle M, Hughes K, Zhao LR. Single-Cell RNA Sequencing Reveals Immunomodulatory Effects of Stem Cell Factor and Granulocyte Colony-Stimulating Factor Treatment in the Brains of Aged APP/PS1 Mice. Biomolecules 2024; 14:827. [PMID: 39062541 PMCID: PMC11275138 DOI: 10.3390/biom14070827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive neurodegeneration and dementia. AD primarily affects older adults with neuropathological changes including amyloid-beta (Aβ) deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) reduces the Aβ load, increases Aβ uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APPswe/PS1dE9 (APP/PS1) mice. However, the mechanisms underlying SCF+G-CSF-enhanced brain repair in aged APP/PS1 mice remain unclear. This study used a transcriptomic approach to identify the potential mechanisms by which SCF+G-CSF treatment modulates microglia and peripheral myeloid cells to mitigate AD pathology in the aged brain. After injections of SCF+G-CSF for 5 consecutive days, single-cell RNA sequencing was performed on CD11b+ cells isolated from the brains of 28-month-old APP/PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF+G-CSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF+G-CSF-induced increase of cerebral CD45high/CD11b+ active phagocytes. SCF+G-CSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. The expression of S100a8 and S100a9 was robustly enhanced following SCF+G-CSF treatment in all CD11b+ cell clusters. Moreover, the topmost genes differentially expressed with SCF+G-CSF treatment were largely upregulated in S100a8/9-positive cells, suggesting a well-conserved transcriptional profile related to SCF+G-CSF treatment in resident and peripherally derived CD11b+ immune cells. This S100a8/9-associated transcriptional profile contained notable genes related to pro-inflammatory and anti-inflammatory responses, neuroprotection, and Aβ plaque inhibition or clearance. Altogether, this study reveals the immunomodulatory effects of SCF+G-CSF treatment in the aged brain with AD pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, 750 E. Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
6
|
Gardner R, Kyle M, Hughes K, Zhao LR. Single cell RNA sequencing reveals immunomodulatory effects of stem cell factor and granulocyte colony-stimulating factor treatment in the brains of aged APP/PS1 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593359. [PMID: 38766064 PMCID: PMC11100789 DOI: 10.1101/2024.05.09.593359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Alzheimers disease leads to progressive neurodegeneration and dementia. Alzheimers disease primarily affects older adults with neuropathological changes including amyloid beta deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor, SCF, and granulocyte colony stimulating factor, GCSF, reduces amyloid beta load, increases amyloid beta uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APP-PS1 mice. However, the mechanisms underlying SCF-GCSF-enhanced brain repair in aged APP-PS1 mice remain unclear. This study used a transcriptomic approach to identify potential mechanisms by which SCF-GCSF treatment modulates microglia and peripheral myeloid cells to mitigate Alzheimers disease pathology in the aged brain. After injections of SCF-GCSF for 5 consecutive days, single cell RNA sequencing was performed on CD11b positive cells isolated from the brains of 28-month-old APP-PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF-GCSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF-GCSF-induced increase of cerebral CD45high-CD11b positive active phagocytes. SCF-GCSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. Expression of S100a8 and S100a9 were robustly enhanced following SCF-GCSF treatment in all CD11b positive cell clusters. Moreover, the topmost genes differentially expressed with SCF-GCSF treatment were largely upregulated in S100a8-S100a9 positive cells, suggesting a well-conserved transcriptional profile related to SCF-GCSF treatment in resident and peripherally derived CD11b positive immune cells. This S100a8-S100a9-associated transcriptional profile contained notable genes related to proinflammatory and antiinflammatory responses, neuroprotection, and amyloid beta plaque inhibition or clearance. Altogether, this study reveals immunomodulatory effects of SCF-GCSF treatment in the aged brain with Alzheimers disease pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
|
7
|
Thomson BR, Schwendinger N, Beckmann K, Gentinetta T, Couto D, Wymann S, Verdon V, Buzzi RM, Akeret K, Kronen PW, Weinberger EM, Held U, Seehusen F, Richter H, Schaer DJ, Hugelshofer M. Haptoglobin Attenuates Cerebrospinal Fluid Hemoglobin-Induced Neurological Deterioration in Sheep. Transl Stroke Res 2024:10.1007/s12975-024-01254-9. [PMID: 38652234 DOI: 10.1007/s12975-024-01254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Secondary brain injury (SBI) occurs with a lag of several days post-bleeding in patients with aneurysmal subarachnoid hemorrhage (aSAH) and is a strong contributor to mortality and long-term morbidity. aSAH-SBI coincides with cell-free hemoglobin (Hb) release into the cerebrospinal fluid. This temporal association and convincing pathophysiological concepts suggest that CSF-Hb could be a targetable trigger of SBI. However, sparse experimental evidence for Hb's neurotoxicity in vivo defines a significant research gap for clinical translation. We modeled the CSF-Hb exposure observed in aSAH patients in conscious sheep, which allowed us to assess neurological functions in a gyrencephalic species. Twelve animals were randomly assigned for 3-day bi-daily intracerebroventricular (ICV) injections of either Hb or Hb combined with the high-affinity Hb scavenger protein haptoglobin (Hb-Hp, CSL888). Repeated CSF sampling confirmed clinically relevant CSF-Hb concentrations. This prolonged CSF-Hb exposure over 3 days resulted in disturbed movement activity, reduced food intake, and impaired observational neuroscores. The Hb-induced neurotoxic effects were significantly attenuated when Hb was administered with equimolar haptoglobin. Preterminal magnetic resonance imaging (MRI) showed no CSF-Hb-specific structural brain alterations. In both groups, histology demonstrated an inflammatory response and revealed enhanced perivascular histiocytic infiltrates in the Hb-Hp group, indicative of adaptive mechanisms. Heme exposure in CSF and iron deposition in the brain were comparable, suggesting comparable clearance efficiency of Hb and Hb-haptoglobin complexes from the intracranial compartment. We identified a neurological phenotype of CSF-Hb toxicity in conscious sheep, which is rather due to neurovascular dysfunction than structural brain injury. Haptoglobin was effective at attenuating CSF-Hb-induced neurological deterioration, supporting its therapeutic potential.
Collapse
Affiliation(s)
- Bart R Thomson
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Nina Schwendinger
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Katrin Beckmann
- Neurology Service, Department of Small Animals, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Thomas Gentinetta
- CSL Biologics Research Centre, Swiss Institute for Translational and Entrepreneurial Medicine, CSL, sitem-insel, Bern, Switzerland
| | - Daniel Couto
- CSL Biologics Research Centre, Swiss Institute for Translational and Entrepreneurial Medicine, CSL, sitem-insel, Bern, Switzerland
| | - Sandra Wymann
- CSL Biologics Research Centre, Swiss Institute for Translational and Entrepreneurial Medicine, CSL, sitem-insel, Bern, Switzerland
| | - Valérie Verdon
- CSL Biologics Research Centre, Swiss Institute for Translational and Entrepreneurial Medicine, CSL, sitem-insel, Bern, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Peter W Kronen
- Veterinary Anaesthesia Services - International, Winterthur, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
| | - Eva M Weinberger
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Ulrike Held
- Department of Biostatistics and Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Frauke Seehusen
- Laboratory of Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Henning Richter
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
- Diagnostic Imaging Research Unit (DIRU), Clinic for Diagnostic Imaging, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland.
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Bandyopadhyay S, Schwendinger N, Jahromi BR, Lad SP, Blackburn S, Wolf S, Bulters D, Galea I, Hugelshofer M. Red Blood Cells in the Cerebrospinal Fluid Compartment After Subarachnoid Haemorrhage: Significance and Emerging Therapeutic Strategies. Transl Stroke Res 2024:10.1007/s12975-024-01238-9. [PMID: 38418755 DOI: 10.1007/s12975-024-01238-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
Subarachnoid haemorrhage (SAH) is a subtype of stroke that predominantly impacts younger individuals. It is associated with high mortality rates and can cause long-term disabilities. This review examines the contribution of the initial blood load and the dynamics of clot clearance to the pathophysiology of SAH and the risk of adverse outcomes. These outcomes include hydrocephalus and delayed cerebral ischaemia (DCI), with a particular focus on the impact of blood located in the cisternal spaces, as opposed to ventricular blood, in the development of DCI. The literature described underscores the prognostic value of haematoma characteristics, such as volume, density, and anatomical location. The limitations of traditional radiographic grading systems are discussed, compared with the more accurate volumetric quantification techniques for predicting patient prognosis. Further, the significance of red blood cells (RBCs) and their breakdown products in secondary brain injury after SAH is explored. The review presents novel interventions designed to accelerate clot clearance or mitigate the effects of toxic byproducts released from erythrolysis in the cerebrospinal fluid following SAH. In conclusion, this review offers deeper insights into the complex dynamics of SAH and discusses the potential pathways available for advancing its management.
Collapse
Affiliation(s)
- Soham Bandyopadhyay
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Nina Schwendinger
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Behnam Rezai Jahromi
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Shivanand P Lad
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Spiros Blackburn
- Department of Neurosurgery, University of Texas Houston Health Science Center, Houston, TX, USA
| | - Stefan Wolf
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Diederik Bulters
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Michael Hugelshofer
- Department of Neurosurgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
9
|
Lauzier DC, Srienc AI, Vellimana AK, Dacey Jr RG, Zipfel GJ. Peripheral macrophages in the development and progression of structural cerebrovascular pathologies. J Cereb Blood Flow Metab 2024; 44:169-191. [PMID: 38000039 PMCID: PMC10993883 DOI: 10.1177/0271678x231217001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 11/26/2023]
Abstract
The human cerebrovascular system is responsible for maintaining neural function through oxygenation, nutrient supply, filtration of toxins, and additional specialized tasks. While the cerebrovascular system has resilience imparted by elaborate redundant collateral circulation from supportive tertiary structures, it is not infallible, and is susceptible to developing structural vascular abnormalities. The causes of this class of structural cerebrovascular diseases can be broadly categorized as 1) intrinsic developmental diseases resulting from genetic or other underlying aberrations (arteriovenous malformations and cavernous malformations) or 2) extrinsic acquired diseases that cause compensatory mechanisms to drive vascular remodeling (aneurysms and arteriovenous fistulae). Cerebrovascular diseases of both types pose significant risks to patients, in some cases leading to death or disability. The drivers of such diseases are extensive, yet inflammation is intimately tied to all of their progressions. Central to this inflammatory hypothesis is the role of peripheral macrophages; targeting this critical cell type may lead to diagnostic and therapeutic advancement in this area. Here, we comprehensively review the role that peripheral macrophages play in cerebrovascular pathogenesis, provide a schema through which macrophage behavior can be understood in cerebrovascular pathologies, and describe emerging diagnostic and therapeutic avenues in this area.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anja I Srienc
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ananth K Vellimana
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ralph G Dacey Jr
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory J Zipfel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Henrich L, Kiessling I, Steimer M, Frase S, Kaiser S, Schallner N. Circadian dependency of microglial heme oxygenase-1 expression and inflammation determine neuronal injury in hemorrhagic stroke. J Inflamm (Lond) 2023; 20:43. [PMID: 38104143 PMCID: PMC10725034 DOI: 10.1186/s12950-023-00371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND The heme oxygenase-1 (HO-1) enzyme pathway is of crucial importance in the removal of toxic blood components and regulation of neuroinflammation following hemorrhagic stroke. Although a circadian pattern dependency in the incidence and severity of hemorrhagic stroke exists, it is unknown whether the activity of the HO-1 system in the context of hemorrhagic injury also exhibits circadian dependency. We hypothesized that the circadian regulation of microglial HO-1 would determine the extent of neuroinflammation and neuronal injury in a murine model of subarachnoid hemorrhage (SAH). METHODS In vitro expression patterns of HO-1 and circadian rhythm genes were analyzed in the microglial BV-2 cell line and primary microglia (PMG) using Western blot and qPCR. PMG isolated from Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice were used to evaluate the role of microglial HO-1. We further investigated the in vivo relevance in a murine subarachnoid hemorrhage (SAH) model using Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice with myeloid cell HO-1 deficiency, inducing SAH at different zeitgeber (ZT) times and analyzing the expression of HO-1 and the circadian control gene Period-2 (Per-2), respectively. Furthermore, we measured the inflammatory cytokine Monocyte Chemoattractant Protein-1 (MCP-1) in the cerebrospinal fluid of SAH patients in correlation with clinical outcome. RESULTS HO-1 baseline expression and response to CO with blood exposure depended on ZT. In vitro expression of circadian control genes was de-synchronized in LyzM-Cre-Hmox1fl/fl PMG and did not respond to exogenous CO exposure. We found that circadian rhythm plays a crucial role in brain damage after SAH. At ZT2, we observed less phagocytic function, more vasospasm and increased microglial activation. CO reduced mortality at ZT12 in HO-1 deficient mice and reduced the difference between ZT2 and ZT12 in the inflammatory response. Induction of MCP-1 in the CSF from SAH patients was time-dependent and correlated with the expression of circadian control genes, SAH severity, functional impairment and delirium. CONCLUSIONS Our data point towards a crucial role for the HO-1 enzyme system and circadian control in neuronal injury after a hemorrhagic stroke.
Collapse
Affiliation(s)
- Luise Henrich
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Iva Kiessling
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matti Steimer
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sibylle Frase
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurology and Neuroscience, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sandra Kaiser
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
11
|
Bandyopadhyay S, Garland P, Gaastra B, Zolnourian A, Bulters D, Galea I. The Haptoglobin Response after Aneurysmal Subarachnoid Haemorrhage. Int J Mol Sci 2023; 24:16922. [PMID: 38069244 PMCID: PMC10707007 DOI: 10.3390/ijms242316922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Haptoglobin is the body's first line of defence against the toxicity of extracellular haemoglobin released following a subarachnoid haemorrhage (SAH). We investigated the haptoglobin response after SAH in cerebrospinal fluid (CSF) and serum. Paired CSF and serum samples from 19 controls and 92 SAH patients were assayed as follows: ultra-performance liquid chromatography for CSF haemoglobin and haptoglobin, immunoassay for serum haptoglobin and multiplexed CSF cytokines, and colorimetry for albumin. There was marked CSF haptoglobin deficiency: 99% of extracellular haemoglobin was unbound. The quotients for both CSF/serum albumin (qAlb) and haptoglobin (qHp) were used to compute the CSF haptoglobin index (qHp/qAlb). CSF from SAH patients had a significantly lower haptoglobin index compared to controls, especially in Haptoglobin-1 allele carriers. Serum haptoglobin levels increased after SAH and were correlated with CSF cytokine levels. Haptoglobin variables were not associated with long-term clinical outcomes post-SAH. We conclude that: (1) intrathecal haptoglobin consumption occurs after SAH, more so in haptoglobin-1 allele carriers; (2) serum haptoglobin is upregulated after SAH, in keeping with the liver acute phase response to central inflammation; (3) haptoglobin in the CSF is so low that any variation is too small for this to affect long-term outcomes, emphasising the potential for therapeutic haptoglobin supplementation.
Collapse
Affiliation(s)
- Soham Bandyopadhyay
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Patrick Garland
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
| | - Ben Gaastra
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Ardalan Zolnourian
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Diederik Bulters
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Ian Galea
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.B.); (P.G.); (B.G.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| |
Collapse
|
12
|
Thomson BR, Gürlek F, Buzzi RM, Schwendinger N, Keller E, Regli L, van Doormaal TP, Schaer DJ, Hugelshofer M, Akeret K. Clinical potential of automated convolutional neural network-based hematoma volumetry after aneurysmal subarachnoid hemorrhage. J Stroke Cerebrovasc Dis 2023; 32:107357. [PMID: 37734180 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
OBJECTIVES Cerebrospinal fluid hemoglobin has been positioned as a potential biomarker and drug target for aneurysmal subarachnoid hemorrhage-related secondary brain injury (SAH-SBI). The maximum amount of hemoglobin, which may be released into the cerebrospinal fluid, is defined by the initial subarachnoid hematoma volume (ISHV). In patients without external ventricular or lumbar drain, there remains an unmet clinical need to predict the risk for SAH-SBI. The aim of this study was to explore automated segmentation of ISHV as a potential surrogate for cerebrospinal fluid hemoglobin to predict SAH-SBI. METHODS This study is based on a retrospective analysis of imaging and clinical data from 220 consecutive patients with aneurysmal subarachnoid hemorrhage collected over a five-year period. 127 annotated initial non-contrast CT scans were used to train and test a convolutional neural network to automatically segment the ISHV in the remaining cohort. Performance was reported in terms of Dice score and intraclass correlation. We characterized the associations between ISHV and baseline cohort characteristics, SAH-SBI, ventriculoperitoneal shunt dependence, functional outcome, and survival. Established clinical (World Federation of Neurosurgical Societies, Hunt & Hess) and radiological (modified Fisher, Barrow Neurological Institute) scores served as references. RESULTS A strong volume agreement (0.73 Dice, range 0.43 - 0.93) and intraclass correlation (0.89, 95% CI, 0.81-0.94) were shown. While ISHV was not associated with the use of antithrombotics or cardiovascular risk factors, there was strong evidence for an association with a lower Glasgow Coma Scale at hospital admission. Aneurysm size and location were not associated with ISHV, but the presence of intracerebral or intraventricular hemorrhage were independently associated with higher ISHV. Despite strong evidence for a positive association between ISHV and SAH-SBI, the discriminatory ability of ISHV for SAH-SBI was insufficient. The discriminatory ability of ISHV was, however, higher regarding ventriculoperitoneal shunt dependence and functional outcome at three-months follow-up. Multivariate survival analysis provided strong evidence for an independent negative association between survival probability and both ISHV and intraventricular hemorrhage. CONCLUSIONS The proposed algorithm demonstrates strong performance in volumetric segmentation of the ISHV on the admission CT. While the discriminatory ability of ISHV for SAH-SBI was similar to established clinical and radiological scores, it showed a high discriminatory ability for ventriculoperitoneal shunt dependence and functional outcome at three-months follow-up.
Collapse
Affiliation(s)
- Bart R Thomson
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland; Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Firat Gürlek
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland; Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Nina Schwendinger
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland
| | - Emanuela Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland; Neurointensive Care Unit, Department of Neurosurgery, and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland
| | - Tristan Pc van Doormaal
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland; Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Translational Neuroscience, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland
| | - Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland.
| |
Collapse
|
13
|
Lauzier DC, Chiang SN, Moran CJ. Etiologies of Brain Arteriovenous Malformation Recurrence: A Focus on Pediatric Disease. Pediatr Neurol 2023; 148:94-100. [PMID: 37690270 DOI: 10.1016/j.pediatrneurol.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Pediatric brain arteriovenous malformations are a major cause of morbidity and mortality, with the harmful effects of this disease compounded by the additional disability-years experienced by children with ruptured or other symptomatic arteriovenous malformations. In addition to the risks shared with their adult counterparts, pediatric patients frequently experience recurrence following radiographic cure, which presents an additional source of morbidity and mortality. Therefore, there is a need to synthesize potential mechanisms contributing to the elevated recurrence risk in the pediatric population and discuss how these translate to practical considerations for managing these patients.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| | - Sarah N Chiang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher J Moran
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
14
|
Warming H, Deinhardt K, Garland P, More J, Bulters D, Galea I, Vargas-Caballero M. Functional effects of haemoglobin can be rescued by haptoglobin in an in vitro model of subarachnoid haemorrhage. J Neurochem 2023; 167:90-103. [PMID: 37702203 DOI: 10.1111/jnc.15936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.
Collapse
Affiliation(s)
- Hannah Warming
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Katrin Deinhardt
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | | | - John More
- Bio Products Laboratory Limited, Elstree, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Mariana Vargas-Caballero
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
15
|
Galea I, Bandyopadhyay S, Bulters D, Humar R, Hugelshofer M, Schaer DJ. Haptoglobin Treatment for Aneurysmal Subarachnoid Hemorrhage: Review and Expert Consensus on Clinical Translation. Stroke 2023; 54:1930-1942. [PMID: 37232189 PMCID: PMC10289236 DOI: 10.1161/strokeaha.123.040205] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/27/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating form of stroke frequently affecting young to middle-aged adults, with an unmet need to improve outcome. This special report focusses on the development of intrathecal haptoglobin supplementation as a treatment by reviewing current knowledge and progress, arriving at a Delphi-based global consensus regarding the pathophysiological role of extracellular hemoglobin and research priorities for clinical translation of hemoglobin-scavenging therapeutics. After aneurysmal subarachnoid hemorrhage, erythrocyte lysis generates cell-free hemoglobin in the cerebrospinal fluid, which is a strong determinant of secondary brain injury and long-term clinical outcome. Haptoglobin is the body's first-line defense against cell-free hemoglobin by binding it irreversibly, preventing translocation of hemoglobin into the brain parenchyma and nitric oxide-sensitive functional compartments of cerebral arteries. In mouse and sheep models, intraventricular administration of haptoglobin reversed hemoglobin-induced clinical, histological, and biochemical features of human aneurysmal subarachnoid hemorrhage. Clinical translation of this strategy imposes unique challenges set by the novel mode of action and the anticipated need for intrathecal drug administration, necessitating early input from stakeholders. Practising clinicians (n=72) and scientific experts (n=28) from 5 continents participated in the Delphi study. Inflammation, microvascular spasm, initial intracranial pressure increase, and disruption of nitric oxide signaling were deemed the most important pathophysiological pathways determining outcome. Cell-free hemoglobin was thought to play an important role mostly in pathways related to iron toxicity, oxidative stress, nitric oxide, and inflammation. While useful, there was consensus that further preclinical work was not a priority, with most believing the field was ready for an early phase trial. The highest research priorities were related to confirming haptoglobin's anticipated safety, individualized versus standard dosing, timing of treatment, pharmacokinetics, pharmacodynamics, and outcome measure selection. These results highlight the need for early phase trials of intracranial haptoglobin for aneurysmal subarachnoid hemorrhage, and the value of early input from clinical disciplines on a global scale during the early stages of clinical translation.
Collapse
Affiliation(s)
- Ian Galea
- Department of Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Hampshire, United Kingdom (I.G., S.B., D.B.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom (I.G., S.B., D.B.)
| | - Soham Bandyopadhyay
- Department of Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Hampshire, United Kingdom (I.G., S.B., D.B.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom (I.G., S.B., D.B.)
| | - Diederik Bulters
- Department of Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Hampshire, United Kingdom (I.G., S.B., D.B.)
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom (I.G., S.B., D.B.)
| | - Rok Humar
- Division of Internal Medicine (R.H., D.J.S.), Universitätsspital and University of Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center (M.H.), Universitätsspital and University of Zurich, Switzerland
| | - Dominik J. Schaer
- Division of Internal Medicine (R.H., D.J.S.), Universitätsspital and University of Zurich, Switzerland
| |
Collapse
|
16
|
Lauzier DC, Jayaraman K, Yuan JY, Diwan D, Vellimana AK, Osbun J, Chatterjee AR, Athiraman U, Dhar R, Zipfel GJ. Early Brain Injury After Subarachnoid Hemorrhage: Incidence and Mechanisms. Stroke 2023; 54:1426-1440. [PMID: 36866673 PMCID: PMC10243167 DOI: 10.1161/strokeaha.122.040072] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Aneurysmal subarachnoid hemorrhage is a devastating condition causing significant morbidity and mortality. While outcomes from subarachnoid hemorrhage have improved in recent years, there continues to be significant interest in identifying therapeutic targets for this disease. In particular, there has been a shift in emphasis toward secondary brain injury that develops in the first 72 hours after subarachnoid hemorrhage. This time period of interest is referred to as the early brain injury period and comprises processes including microcirculatory dysfunction, blood-brain-barrier breakdown, neuroinflammation, cerebral edema, oxidative cascades, and neuronal death. Advances in our understanding of the mechanisms defining the early brain injury period have been accompanied by improved imaging and nonimaging biomarkers for identifying early brain injury, leading to the recognition of an elevated clinical incidence of early brain injury compared with prior estimates. With the frequency, impact, and mechanisms of early brain injury better defined, there is a need to review the literature in this area to guide preclinical and clinical study.
Collapse
Affiliation(s)
- David C. Lauzier
- Department of Neurological Surgery, Washington University School of Medicine
| | - Keshav Jayaraman
- Department of Neurological Surgery, Washington University School of Medicine
| | - Jane Y. Yuan
- Department of Neurological Surgery, Washington University School of Medicine
| | - Deepti Diwan
- Department of Neurological Surgery, Washington University School of Medicine
| | - Ananth K. Vellimana
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | - Joshua Osbun
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | - Arindam R. Chatterjee
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | | | - Rajat Dhar
- Department of Neurology, Washington University School of Medicine
| | - Gregory J. Zipfel
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
| |
Collapse
|
17
|
Tartara F, Montalbetti A, Crobeddu E, Armocida D, Tavazzi E, Cardia A, Cenzato M, Boeris D, Garbossa D, Cofano F. Compartmental Cerebrospinal Fluid Events Occurring after Subarachnoid Hemorrhage: An "Heparin Oriented" Systematic Review. Int J Mol Sci 2023; 24:7832. [PMID: 37175544 PMCID: PMC10178276 DOI: 10.3390/ijms24097832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) represents a severe acute event with high morbidity and mortality due to the development of early brain injury (EBI), secondary delayed cerebral ischemia (DCI), and shunt-related hydrocephalus. Secondary events (SSE) such as neuroinflammation, vasospasm, excitotoxicity, blood-brain barrier disruption, oxidative cascade, and neuronal apoptosis are related to DCI. Despite improvement in management strategies and therapeutic protocols, surviving patients frequently present neurological deficits with neurocognitive impairment. The aim of this paper is to offer to clinicians a practical review of the actually documented pathophysiological events following subarachnoid hemorrhage. To reach our goal we performed a literature review analyzing reported studies regarding the mediators involved in the pathophysiological events following SAH occurring in the cerebrospinal fluid (CSF) (hemoglobin degradation products, platelets, complement, cytokines, chemokines, leucocytes, endothelin-1, NO-synthase, osteopontin, matricellular proteins, blood-brain barrier disruption, microglia polarization). The cascade of pathophysiological events secondary to SAH is very complex and involves several interconnected, but also distinct pathways. The identification of single therapeutical targets or specific pharmacological agents may be a limited strategy able to block only selective pathophysiological paths, but not the global evolution of SAH-related events. We report furthermore on the role of heparin in SAH management and discuss the rationale for use of intrathecal heparin as a pleiotropic therapeutical agent. The combination of the anticoagulant effect and the ability to interfere with SSE theoretically make heparin a very interesting molecule for SAH management.
Collapse
Affiliation(s)
- Fulvio Tartara
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Montalbetti
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Emanuela Crobeddu
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Daniele Armocida
- A.U.O. Policlinico Umberto I, Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00185 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Eleonora Tavazzi
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Cardia
- Department of Neurosurgery, Neurocenter of Southern Switzerland, EOC, 6900 Lugano, Switzerland
| | - Marco Cenzato
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Davide Boeris
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Diego Garbossa
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| | - Fabio Cofano
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| |
Collapse
|
18
|
Sulimai N, Brown J, Lominadze D. Vascular Effects on Cerebrovascular Permeability and Neurodegeneration. Biomolecules 2023; 13:648. [PMID: 37189395 PMCID: PMC10136045 DOI: 10.3390/biom13040648] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Neurons and glial cells in the brain are protected by the blood brain barrier (BBB). The local regulation of blood flow is determined by neurons and signal conducting cells called astrocytes. Although alterations in neurons and glial cells affect the function of neurons, the majority of effects are coming from other cells and organs of the body. Although it seems obvious that effects beginning in brain vasculature would play an important role in the development of various neuroinflammatory and neurodegenerative pathologies, significant interest has only been directed to the possible mechanisms involved in the development of vascular cognitive impairment and dementia (VCID) for the last decade. Presently, the National Institute of Neurological Disorders and Stroke applies considerable attention toward research related to VCID and vascular impairments during Alzheimer's disease. Thus, any changes in cerebral vessels, such as in blood flow, thrombogenesis, permeability, or others, which affect the proper vasculo-neuronal connection and interaction and result in neuronal degeneration that leads to memory decline should be considered as a subject of investigation under the VCID category. Out of several vascular effects that can trigger neurodegeneration, changes in cerebrovascular permeability seem to result in the most devastating effects. The present review emphasizes the importance of changes in the BBB and possible mechanisms primarily involving fibrinogen in the development and/or progression of neuroinflammatory and neurodegenerative diseases resulting in memory decline.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA; (N.S.); (J.B.)
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
19
|
Thomson BR, Richter H, Akeret K, Buzzi RM, Anagnostakou V, van Niftrik CHB, Schwendinger N, Kulcsar Z, Kronen PW, Regli L, Fierstra J, Schaer DJ, Hugelshofer M. Blood oxygenation-level dependent cerebrovascular reactivity imaging as strategy to monitor CSF-hemoglobin toxicity. J Stroke Cerebrovasc Dis 2023; 32:106985. [PMID: 36640721 DOI: 10.1016/j.jstrokecerebrovasdis.2023.106985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVES Cell-free hemoglobin in the cerebrospinal fluid (CSF-Hb) may be one of the main drivers of secondary brain injury after aneurysmal subarachnoid hemorrhage (aSAH). Haptoglobin scavenging of CSF-Hb has been shown to mitigate cerebrovascular disruption. Using digital subtraction angiography (DSA) and blood oxygenation-level dependent cerebrovascular reactivity imaging (BOLD-CVR) the aim was to assess the acute toxic effect of CSF-Hb on cerebral blood flow and autoregulation, as well as to test the protective effects of haptoglobin. METHODS DSA imaging was performed in eight anesthetized and ventilated sheep (mean weight: 80.4 kg) at baseline, 15, 30, 45 and 60 minutes after infusion of hemoglobin (Hb) or co-infusion with haptoglobin (Hb:Haptoglobin) into the left lateral ventricle. Additionally, 10 ventilated sheep (mean weight: 79.8 kg) underwent BOLD-CVR imaging to assess the cerebrovascular reserve capacity. RESULTS DSA imaging did not show a difference in mean transit time or cerebral blood flow. Whole-brain BOLD-CVR compared to baseline decreased more in the Hb group after 15 minutes (Hb vs Hb:Haptoglobin: -0.03 ± 0.01 vs -0.01 ± 0.02) and remained diminished compared to Hb:Haptoglobin group after 30 minutes (Hb vs Hb:Haptoglobin: -0.03 ± 0.01 vs 0.0 ± 0.01), 45 minutes (Hb vs Hb:Haptoglobin: -0.03 ± 0.01 vs 0.01 ± 0.02) and 60 minutes (Hb vs Hb:Haptoglobin: -0.03 ± 0.02 vs 0.01 ± 0.01). CONCLUSION It is demonstrated that CSF-Hb toxicity leads to rapid cerebrovascular reactivity impairment, which is blunted by haptoglobin co-infusion. BOLD-CVR may therefore be further evaluated as a monitoring strategy for CSF-Hb toxicity after aSAH.
Collapse
Affiliation(s)
- Bart R Thomson
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland; Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Henning Richter
- Clinic for Diagnostic Imaging, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland; Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
| | - Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Vania Anagnostakou
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland; Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christiaan H B van Niftrik
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Nina Schwendinger
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Zsolt Kulcsar
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland; Department of Neuroradiology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Peter W Kronen
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland; Veterinary Anaesthesia Services - International, Winterthur, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jorn Fierstra
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland; Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland.
| |
Collapse
|
20
|
Hopkins K, Price B, Ziogas J, Adamides A, Mangum J. Comparative proteomic analysis of ventricular and cisternal cerebrospinal fluid in haemorrhagic stroke patients. J Clin Neurosci 2023; 107:84-90. [PMID: 36525746 DOI: 10.1016/j.jocn.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Analysis of cerebrospinal fluid (CSF) using mass spectrometry is a relatively novel analytical tool, and comparisons of ventricular and cisternal proteomes are yet to be performed. This may have implications for clinical medicine, particularly in demonstrating continuity of the ventricular system with preserved flow in the presence of ventricular blood. Other uses include the identification of novel biomarkers, including for diagnosis of subarachnoid haemorrhage and of aetiology. The primary objective was therefore to characterise and compare the proteomes of ventricular and CSF after haemorrhagic stroke. METHODS Paired CSF samples were prospectively collected from the optico-carotid cistern and the frontal horn of the lateral ventricle at the time of craniotomy and clipping in 8 patients with haemorrhagic stroke. Six patients had an aneurysmal subarachnoid haemorrhage (aSAH) from a ruptured saccular aneurysm, one patient had an aSAH after rupture of a mycotic aneurysm and one patient had a spontaneous intracerebral haemorrhage (IPH) with an adjacent unruptured saccular aneurysm. Samples were processed and proteins identified and quantified using data-dependent liquid chromatography tandem mass spectrometry (DDA LC-MSMS). RESULTS There was no systematic difference between the cisternal and ventricular proteomes. However, blinded principal component analysis (PCA) of the cisternal and ventricular samples separated patients according to pathophysiology. Additionally CSF D-Dimer levels were not detected in the IPH patient but were reliably measured in aSAH patients. CONCLUSIONS Ventricular CSF is representative of cisternal CSF after aSAH. CSF proteomic PCA analysis can distinguish between haemorrhage types. CSF D-dimer levels may represent a novel diagnostic marker for aSAH. Label free DDA LC-MSMS CSF analysis may inform possible biomarkers.
Collapse
Affiliation(s)
- Katherine Hopkins
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin Price
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Melbourne, VIC, Australia.
| | - James Ziogas
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC, Australia
| | - Alexios Adamides
- Department of Neurosurgery, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Jonathan Mangum
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Vallelian F, Buehler PW, Schaer DJ. Hemolysis, free hemoglobin toxicity, and scavenger protein therapeutics. Blood 2022; 140:1837-1844. [PMID: 35660854 PMCID: PMC10653008 DOI: 10.1182/blood.2022015596] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/27/2022] [Indexed: 11/20/2022] Open
Abstract
During hemolysis, erythrophagocytes dispose damaged red blood cells. This prevents the extracellular release of hemoglobin, detoxifies heme, and recycles iron in a linked metabolic pathway. Complementary to this process, haptoglobin and hemopexin scavenge and shuttle the red blood cell toxins hemoglobin and heme to cellular clearance. Pathological hemolysis outpaces macrophage capacity and scavenger synthesis across a diversity of diseases. This imbalance leads to hemoglobin-driven disease progression. To meet a void in treatment options, scavenger protein-based therapeutics are in clinical development.
Collapse
Affiliation(s)
- Florence Vallelian
- Division of Internal Medicine, University Hospital, University of Zurich, Zurich, Switzerland
| | - Paul W. Buehler
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
- Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD
| | - Dominik J. Schaer
- Division of Internal Medicine, University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Mahmoodkhani M, Sharafi M, Sourani A, Tehrani DS. Half-Saline Versus Normal-Saline as Irrigation Solutions in Burr Hole Craniostomy to Treat Chronic Subdural Hematomata: A Randomized Clinical Trial. Korean J Neurotrauma 2022; 18:221-229. [PMID: 36381457 PMCID: PMC9634318 DOI: 10.13004/kjnt.2022.18.e47] [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: 06/16/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 06/16/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the efficacy and safety of half-saline (HS) serum as an irrigation solution in chronic subdural hematoma (CSDH) surgery using the burr hole craniostomy (BHC) technique. METHODS This randomized clinical trial was conducted in university hospital referral centers from 2020 to 2021. Sixty-three patients with CSDH eligible for BHC were primarily enrolled. Two patients were excluded because of concurrent stroke. Sixty-one patients were randomly allocated into case (HS=30) and control (normal-saline [NS]=31) groups. HS was used to irrigate the hematoma in the case group and NS was used in the control group. The patients were followed-up. Clinical variables including demographic and medical findings, postoperative computed tomography findings, postoperative complications, hospitalization period, recurrence rate, and functional status measured by the Barthel type B index were recorded. RESULTS Forty-six of 61 patients were male (75.4%), and the patients' mean age was 65.4±16.9 years, with equal distribution between the 2 groups. Postoperative effusion and postoperative hospital stay duration were significantly lower in the HS group than in the NS group (p=0.002 and 0.033, respectively). The postoperative recurrence within 3 months in both groups was approximately equal (6.6%). In terms of functional outcomes and postoperative complications, HS showed similar results to those of NS. CONCLUSION HS as an irrigation fluid in BHC effectively reduced postoperative effusion and hospital stay duration without considerable complications. TRIAL REGISTRATION Iranian Registry of Clinical Trials Identifier: IRCT20200608047688N1.
Collapse
Affiliation(s)
- Mehdi Mahmoodkhani
- Department of Neurosurgery, Kashani Hospital, School of Medicine, Isfahan University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharafi
- Department of Neurosurgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arman Sourani
- Department of Neurosurgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
23
|
Zeineddine HA, Honarpisheh P, McBride D, Pandit PKT, Dienel A, Hong SH, Grotta J, Blackburn S. Targeting Hemoglobin to Reduce Delayed Cerebral Ischemia After Subarachnoid Hemorrhage. Transl Stroke Res 2022; 13:725-735. [PMID: 35157256 PMCID: PMC9375776 DOI: 10.1007/s12975-022-00995-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
Abstract
Delayed cerebral ischemia (DCI) continues to be a sequela of aneurysmal subarachnoid hemorrhage (aSAH) that carries significant morbidity and mortality. Aside from nimodipine, no therapeutic agents are available to reduce the incidence of DCI. Pathophysiologic mechanisms contributing to DCI are poorly understood, but accumulating evidence over the years implicates several factors. Those have included microvessel vasoconstriction, microthrombosis, oxidative tissue damage, and cortical spreading depolarization as well as large vessel vasospasm. Common to these processes is red blood cell leakage into the cerebrospinal fluids (CSF) and subsequent lysis which releases hemoglobin, a central instigator in these events. This has led to the hypothesis that early blood removal may improve clinical outcome and reduce DCI. This paper will provide a narrative review of the evidence of hemoglobin as an instigator of DCI. It will also elaborate on available human data that discuss blood clearance and CSF drainage as a treatment of DCI. Finally, we will address a recent novel device that is currently being tested, the Neurapheresis CSF Management System™. This is an automated dual-lumen lumbar drainage system that has an option to filter CSF and return it to the patient.
Collapse
Affiliation(s)
- Hussein A Zeineddine
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA
| | - Pedram Honarpisheh
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA
| | - Devin McBride
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA
| | - Peeyush Kumar Thankamani Pandit
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA
| | - Ari Dienel
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA
| | - Sung-Ha Hong
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA
| | - James Grotta
- Clinical Innovation and Research Institute, Memorial Hermann Hospital-Texas Medical Center, Houston, TX, USA
| | - Spiros Blackburn
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin Street, Suite 2800, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Gidda R, Bandyopadhyay S, Peter N, Lakhoo K. Decompressive Craniectomy for Pediatric Traumatic Brain Injury in Low-and-Middle Income and High Income Countries. World Neurosurg 2022; 166:251-260.e1. [PMID: 35872132 DOI: 10.1016/j.wneu.2022.07.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 12/15/2022]
Abstract
Traumatic brain injury is one of the leading causes of mortality and morbidity in children worldwide. In severe cases, high intracranial pressure is the most frequent cause of death. When first-line medical management fails, the neurosurgical procedure of decompressive craniectomy (DC) has been proposed for controlling intracranial pressure and improving the long-term outcomes for children with severe traumatic brain injury. However, the use of this procedure is controversial. The evidence from clinical trials shows some promise for the use of DC as an effective second-line treatment. However, it is limited by conflicting trial results, a lack of trials, and a high risk of bias. Furthermore, most research comes from retrospective observational studies and case series. This narrative review considers the strength of evidence for the use of DC in both a high income country and low-and-middle income country setting and examine how we can improve study design to better assess the efficacy of this procedure and increase the clinical translatability of results to centers worldwide. Specifically, we argue for a need for further studies with higher pediatric participant numbers, multicenter collaboration, and the use of a more consistent methodology to enable comparability of results among settings.
Collapse
Affiliation(s)
- Ryan Gidda
- Oxford University Global Surgery Group, Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.
| | - Soham Bandyopadhyay
- Oxford University Global Surgery Group, Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Noel Peter
- Oxford University Global Surgery Group, Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Kokila Lakhoo
- Oxford University Global Surgery Group, Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Bücker P, Buzzi RM, Akeret K, Mosberger L, Richter H, Sperling M, Hugelshofer M, Schaer DJ, Karst U. A model to visualize the fate of iron after intracranial hemorrhage using isotopic tracers and elemental bioimaging. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6652217. [PMID: 35906878 DOI: 10.1093/mtomcs/mfac057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022]
Abstract
Hemoglobin-iron is a red-blood-cell toxin contributing to secondary brain injury after intracranial bleeding. We present a model to visualize an intracerebral hematoma and secondary hemoglobin-iron distribution by detecting 58Fe-labeled hemoglobin (Hb) with laser ablation-inductively coupled plasma-mass spectrometry on mouse brain cryosections after stereotactic whole blood injection for different time periods. The generation of 58Fe-enriched blood and decisive steps in the acute hemorrhage formation and evolution was evaluated. The model allows to visualize and quantify 58Fe with high spatial resolution and striking signal-to-noise ratio. Script-based evaluation of the delocalization-depth revealed ongoing 58Fe delocalization in the brain even six days after hematoma induction. Collectively, the model can quantify the distribution of Hb-derived iron post-bleeding, providing a methodological framework to study the pathophysiological basis of cell-free Hb toxicity in hemorrhagic stroke.
Collapse
Affiliation(s)
- Patrick Bücker
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Leila Mosberger
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Henning Richter
- Diagnostic Imaging Research Unit, Clinic for Diagnostic Imaging, University of Zurich, Zurich, Switzerland
| | - Michael Sperling
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| |
Collapse
|
26
|
Akeret K, Buzzi RM, Saxenhofer M, Bieri K, Chiavi D, Thomson BR, Grüttner-Durmaz M, Schwendinger N, Humar R, Regli L, van Doormaal TPC, Held U, Keller E, Hugelshofer M, Schaer DJ. The HeMoVal study protocol: a prospective international multicenter cohort study to validate cerebrospinal fluid hemoglobin as a monitoring biomarker for aneurysmal subarachnoid hemorrhage related secondary brain injury. BMC Neurol 2022; 22:267. [PMID: 35850705 PMCID: PMC9290286 DOI: 10.1186/s12883-022-02789-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Preclinical studies provided a strong rationale for a pathophysiological link between cell-free hemoglobin in the cerebrospinal fluid (CSF-Hb) and secondary brain injury after subarachnoid hemorrhage (SAH-SBI). In a single-center prospective observational clinical study, external ventricular drain (EVD) based CSF-Hb proved to be a promising biomarker to monitor for SAH-SBI. The primary objective of the HeMoVal study is to prospectively validate the association between EVD based CSF-Hb and SAH-SBI during the first 14 days post-SAH. Secondary objectives include the assessment of the discrimination ability of EVD based CSF-Hb for SAH-SBI and the definition of a clinically relevant range of EVD based CSF-Hb toxicity. In addition, lumbar drain (LD) based CSF-Hb will be assessed for its association with and discrimination ability for SAH-SBI. METHODS HeMoVal is a prospective international multicenter observational cohort study. Adult patients admitted with aneurysmal subarachnoid hemorrhage (aSAH) are eligible. While all patients with aSAH are included, we target a sample size of 250 patients with EVD within the first 14 day after aSAH. Epidemiologic and disease-specific baseline measures are assessed at the time of study inclusion. In patients with EVD or LD, each day during the first 14 days post-SAH, 2 ml of CSF will be sampled in the morning, followed by assessment of the patients for SAH-SBI, co-interventions, and complications in the afternoon. After 3 months, a clinical follow-up will be performed. For statistical analysis, the cohort will be stratified into an EVD, LD and full cohort. The primary analysis will quantify the strength of association between EVD based CSF-Hb and SAH-SBI in the EVD cohort based on a generalized additive model. Secondary analyses include the strength of association between LD based CSF-Hb and SAH-SBI in the LD cohort based on a generalized additive model, as well as the discrimination ability of CSF-Hb for SAH-SBI based on receiver operating characteristic (ROC) analyses. DISCUSSION We hypothesize that this study will validate the value of CSF-Hb as a biomarker to monitor for SAH-SBI. In addition, the results of this study will provide the potential base to define an intervention threshold for future studies targeting CSF-Hb toxicity after aSAH. STUDY REGISTRATION ClinicalTrials.gov Identifier NCT04998370 . Date of registration: August 10, 2021.
Collapse
Affiliation(s)
- Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | | | | | - Deborah Chiavi
- Department of Biostatistics at Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Bart R Thomson
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Manuela Grüttner-Durmaz
- Clinical Trials Center - Research Ward (CTC-RW), University Hospital Zurich, Zurich, Switzerland
| | - Nina Schwendinger
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Rok Humar
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Tristan P C van Doormaal
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland
- Department of Neurology and Neurosurgery, University Medical Center, Utrecht, The Netherlands
| | - Ulrike Held
- Department of Biostatistics at Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Emanuela Keller
- Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital and University of Zurich, Zurich, Switzerland.
| | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS 2022; 19:29. [PMID: 35410231 PMCID: PMC8996682 DOI: 10.1186/s12987-022-00312-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The response of the blood-brain barrier (BBB) following a stroke, including subarachnoid hemorrhage (SAH), has been studied extensively. The main components of this reaction are endothelial cells, pericytes, and astrocytes that affect microglia, neurons, and vascular smooth muscle cells. SAH induces alterations in individual BBB cells, leading to brain homeostasis disruption. Recent experiments have uncovered many pathophysiological cascades affecting the BBB following SAH. Targeting some of these pathways is important for restoring brain function following SAH. BBB injury occurs immediately after SAH and has long-lasting consequences, but most changes in the pathophysiological cascades occur in the first few days following SAH. These changes determine the development of early brain injury as well as delayed cerebral ischemia. SAH-induced neuroprotection also plays an important role and weakens the negative impact of SAH. Supporting some of these beneficial cascades while attenuating the major pathophysiological pathways might be decisive in inhibiting the negative impact of bleeding in the subarachnoid space. In this review, we attempt a comprehensive overview of the current knowledge on the molecular and cellular changes in the BBB following SAH and their possible modulation by various drugs and substances.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Klaudia Lakatosová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
28
|
Akeret K, Hugelshofer M, Schaer DJ, Buzzi RM. Spatial transcriptome data from coronal mouse brain sections after striatal injection of heme and heme-hemopexin. Data Brief 2022; 41:107866. [PMID: 35141374 PMCID: PMC8814302 DOI: 10.1016/j.dib.2022.107866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/04/2022] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
Hemorrhagic stroke is a major cause of morbidity and mortality worldwide. Secondary mechanisms of brain injury adversely affect functional outcome in patients after intracranial hemorrhage. Potential drivers of intracranial hemorrhage-related secondary brain injury are hemoglobin and its downstream degradation products released from lysed red blood cells, such as free heme. We established a mouse model with stereotactic striatal injection of heme-albumin to gain insights into the toxicity mechanisms of free heme in the brain and assess the therapeutic potential of heme binding and biochemical neutralization by hemopexin. We defined the dose-dependent transcriptional effect of heme or heme-hemopexin exposure 24 h after injection by spatial transcriptome analysis of lesion-centered coronal cryosections. The spatial transcriptome was interpreted in a multimodal approach along with histology, magnetic resonance imaging, and behavioral data and reported in the associated research article “Spatial transcriptome analysis defines heme as a hemopexin-targetable inflammatoxin in the brain” [1]. The spatially resolved transcriptome dataset made available here is intended for continued analysis of free heme toxicity in the brain, which is of potential pathophysiological and therapeutic significance in the context of a wide range of neurovascular and neurodegenerative diseases.
Collapse
|
29
|
Grossen AA, Ernst GL, Bauer AM. Update on intrathecal management of cerebral vasospasm: a systematic review and meta-analysis. Neurosurg Focus 2022; 52:E10. [PMID: 35231885 DOI: 10.3171/2021.12.focus21629] [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] [Received: 11/01/2021] [Accepted: 12/22/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Aneurysmal subarachnoid hemorrhage (aSAH) accounts for a relatively small portion of strokes but has the potential to cause permanent neurological deficits. Vasospasm with delayed ischemic neurological deficit is thought to be responsible for much of the morbidity associated with aSAH. This has illuminated some treatment options that have the potential to target specific components of the vasospasm cascade. Intrathecal management via lumbar drain (LD) or external ventricular drain (EVD) offers unique advantages in this patient population. The aim of this review was to provide an update on intrathecal vasospasm treatments, emphasizing the need for larger-scale trials and updated protocols using data-driven evidence. METHODS A search of PubMed, Ovid MEDLINE, and Cochrane databases included the search terms (subarachnoid hemorrhage) AND (vasospasm OR delayed cerebral ischemia) AND (intrathecal OR intraventricular OR lumbar drain OR lumbar catheter) for 2010 to the present. Next, a meta-analysis was performed of select therapeutic regimens. The primary endpoints of analysis were vasospasm, delayed cerebral ischemia (DCI), cerebral infarction, and functional outcome. RESULTS Twenty-nine studies were included in the analysis. There were 10 studies in which CSF drainage was the primary experimental group. Calcium channel antagonists were the focus of 7 studies. Fibrinolytics and other vasodilators were each examined in 6 studies. The meta-analysis included studies examining CSF drainage via LD (n = 4), tissue plasminogen activator in addition to EVD (n = 3), intraventricular nimodipine (n = 2), and cisternal magnesium (n = 2). Results showed that intraventricular nimodipine decreased vasospasm (OR 0.59, 95% CI 0.37-0.94; p = 0.03). Therapies that significantly reduced DCI were CSF drainage via LD (OR 0.47, 95% CI 0.25-0.88; p = 0.02) and cisternal magnesium (OR 0.27, 95% CI 0.07-1.02; p = 0.05). CSF drainage via LD was also found to significantly reduce the incidence of cerebral infarction (OR 0.35, 95% 0.24-0.51; p < 0.001). Lastly, functional outcome was significantly better in patients who received CSF drainage via LD (OR 2.42, 95% CI 1.39-4.21; p = 0.002). CONCLUSIONS The authors' results showed that intrathecal therapy is a safe and feasible option following aSAH. It has been shown to attenuate cerebral vasospasm, reduce the incidence of DCI, and improve clinical outcome. The authors support the use of intrathecal management in the prevention and rescue management of cerebral vasospasm. More randomized controlled trials are warranted to determine the best combination of pharmaceutical agents and administration route in order to formulate a standardized treatment approach.
Collapse
|
30
|
Galea I, Durnford A, Glazier J, Mitchell S, Kohli S, Foulkes L, Norman J, Darekar A, Love S, Bulters DO, Nicoll JAR, Boche D. Iron Deposition in the Brain After Aneurysmal Subarachnoid Hemorrhage. Stroke 2022; 53:1633-1642. [PMID: 35196874 DOI: 10.1161/strokeaha.121.036645] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND After aneurysmal subarachnoid hemorrhage (SAH), thrombus forms over the cerebral cortex and releases hemoglobin. When extracellular, hemoglobin is toxic to neurones. High local hemoglobin concentration overwhelms the clearance capacity of macrophages expressing the hemoglobin-haptoglobin scavenger receptor CD163. We hypothesized that iron is deposited in the cortex after SAH and would associate with outcome. METHODS Two complementary cross-sectional studies were conducted. Postmortem brain tissue from 39 SAH (mean postictal interval of 9 days) and 22 control cases was studied with Perls' staining for iron and immunolabeling for CD163, ADAM17 (a disintegrin and metallopeptidase domain 17), CD68, and Iba1 (ionized calcium binding adaptor molecule 1). In parallel, to study the persistence of cortical iron and its relationship to clinical outcome, we conducted a susceptibility-weighted imaging study of 21 SAH patients 6 months postictus and 10 control individuals. RESULTS In brain tissue from patients dying soon after SAH, the distribution of iron deposition followed a gradient that diminished with distance from the brain surface. Iron was located intracellularly (mainly in macrophages, and occasionally in microglia, neurones, and glial cells) and extracellularly. Microglial activation and motility markers were increased after SAH, with a similar inward diminishing gradient. In controls, there was a positive correlation between CD163 and iron, which was lost after SAH. In SAH survivors, iron-sensitive imaging 6 months post-SAH confirmed persistence of cortical iron, related to the size and location of the blood clot immediately after SAH, and associated with cognitive outcome. CONCLUSIONS After SAH, iron deposits in the cortical gray matter in a pattern that reflects proximity to the brain surface and thrombus and is related to cognitive outcome. These observations support therapeutic manoeuvres which prevent the permeation of hemoglobin into the cortex after SAH.
Collapse
Affiliation(s)
- Ian Galea
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.)
| | - Andrew Durnford
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.).,Wessex Neurological Centre (A. Durnford, D.O.B.), University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - James Glazier
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.)
| | - Sophie Mitchell
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.)
| | - Suraj Kohli
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.)
| | | | - Jeanette Norman
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.)
| | - Angela Darekar
- Medical Physics (A. Darekar), University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Seth Love
- Dementia Research Group, Bristol Medical School, University of Bristol, United Kingdom (S.L.)
| | - Diederik O Bulters
- Wessex Neurological Centre (A. Durnford, D.O.B.), University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - James A R Nicoll
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.).,Department of Cellular Pathology (J.A.R.N.), University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Delphine Boche
- Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, United Kingdom (I.G., A. Durnford, J.G., S.M., S.K., J.N., J.A.R.N., D.B.)
| |
Collapse
|
31
|
Akeret K, Buzzi RM, Schaer CA, Thomson BR, Vallelian F, Wang S, Willms J, Sebök M, Held U, Deuel JW, Humar R, Regli L, Keller E, Hugelshofer M, Schaer DJ. Cerebrospinal fluid hemoglobin drives subarachnoid hemorrhage-related secondary brain injury. J Cereb Blood Flow Metab 2021; 41:3000-3015. [PMID: 34102922 PMCID: PMC8545037 DOI: 10.1177/0271678x211020629] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Secondary brain injury after aneurysmal subarachnoid hemorrhage (SAH-SBI) contributes to poor outcomes in patients after rupture of an intracranial aneurysm. The lack of diagnostic biomarkers and novel drug targets represent an unmet need. The aim of this study was to investigate the clinical and pathophysiological association between cerebrospinal fluid hemoglobin (CSF-Hb) and SAH-SBI. In a cohort of 47 patients, we collected daily CSF-samples within 14 days after aneurysm rupture. There was very strong evidence for a positive association between spectrophotometrically determined CSF-Hb and SAH-SBI. The accuracy of CSF-Hb to monitor for SAH-SBI markedly exceeded that of established methods (AUC: 0.89 [0.85-0.92]). Temporal proteome analysis revealed erythrolysis accompanied by an adaptive macrophage response as the two dominant biological processes in the CSF-space after aneurysm rupture. Ex-vivo experiments on the vasoconstrictive and oxidative potential of Hb revealed critical inflection points overlapping CSF-Hb thresholds in patients with SAH-SBI. Selective depletion and in-solution neutralization by haptoglobin or hemopexin efficiently attenuated the vasoconstrictive and lipid peroxidation activities of CSF-Hb. Collectively, the clinical association between high CSF-Hb levels and SAH-SBI, the underlying pathophysiological rationale, and the favorable effects of haptoglobin and hemopexin in ex-vivo experiments position CSF-Hb as a highly attractive biomarker and potential drug target.
Collapse
Affiliation(s)
- Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Christian A Schaer
- Department of Anesthesiology, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Bart R Thomson
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Sophie Wang
- Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Jan Willms
- Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Martina Sebök
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Ulrike Held
- Epidemiology, Biostatistics and Prevention Institute, Department of Biostatistics, University of Zurich; Zurich, Switzerland
| | - Jeremy W Deuel
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Rok Humar
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Emanuela Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland.,Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| |
Collapse
|
32
|
Wu F, Liu Z, Li G, Zhou L, Huang K, Wu Z, Zhan R, Shen J. Inflammation and Oxidative Stress: Potential Targets for Improving Prognosis After Subarachnoid Hemorrhage. Front Cell Neurosci 2021; 15:739506. [PMID: 34630043 PMCID: PMC8497759 DOI: 10.3389/fncel.2021.739506] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) has a high mortality rate and causes long-term disability in many patients, often associated with cognitive impairment. However, the pathogenesis of delayed brain dysfunction after SAH is not fully understood. A growing body of evidence suggests that neuroinflammation and oxidative stress play a negative role in neurofunctional deficits. Red blood cells and hemoglobin, immune cells, proinflammatory cytokines, and peroxidases are directly or indirectly involved in the regulation of neuroinflammation and oxidative stress in the central nervous system after SAH. This review explores the role of various cellular and acellular components in secondary inflammation and oxidative stress after SAH, and aims to provide new ideas for clinical treatment to improve the prognosis of SAH.
Collapse
Affiliation(s)
- Fan Wu
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zongchi Liu
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ganglei Li
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lihui Zhou
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kaiyuan Huang
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhanxiong Wu
- College of Electronics and Information, Hangzhou Dianzi University, Hangzhou, China
| | - Renya Zhan
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Shen
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
33
|
Hunter M, Spiller KJ, Dominique MA, Xu H, Hunter FW, Fang TC, Canter RG, Roberts CJ, Ransohoff RM, Trojanowski JQ, Lee VMY. Microglial transcriptome analysis in the rNLS8 mouse model of TDP-43 proteinopathy reveals discrete expression profiles associated with neurodegenerative progression and recovery. Acta Neuropathol Commun 2021; 9:140. [PMID: 34412701 PMCID: PMC8377972 DOI: 10.1186/s40478-021-01239-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022] Open
Abstract
The microglial reaction is a hallmark of neurodegenerative conditions, and elements thereof may exert differential effects on disease progression, either worsening or ameliorating severity. In amyotrophic lateral sclerosis (ALS), a syndrome characterized by cytoplasmic aggregation of TDP-43 protein and atrophy of motor neurons in the cortex and spinal cord, the transcriptomic signatures of microglia during disease progression are incompletely understood. Here, we performed longitudinal RNAseq analysis of cortical and spinal cord microglia from rNLS8 mice, in which doxycycline-regulatable expression of human TDP-43 (hTDP-43) in the cytoplasm of neurons recapitulates many features of ALS. Transgene suppression in rNLS8 mice leads to functional, anatomical and electrophysiological resolution that is dependent on a microglial reaction that is concurrent with recovery rather than disease onset. We identified basal differences between the gene expression profiles of microglia dependent on localization in spinal cord or cortex. Microglia subjected to chronic hTDP-43 overexpression demonstrated transcriptomic changes in both locations. We noted strong upregulation of Apoe, Axl, Cd63, Clec7a, Csf1, Cst7, Igf1, Itgax, Lgals3, Lilrb4, Lpl and Spp1 during late disease and recovery. Importantly, we identified a distinct suite of differentially expressed genes associated with each phase of disease progression and recovery. Differentially expressed genes were associated with chemotaxis, phagocytosis, inflammation, and production of neuroprotective factors. These data provide new insights into the microglial reaction in TDP-43 proteinopathy. Genes differentially expressed during progression and recovery may provide insight into a unique instance in which the microglial reaction promotes functional recovery after neuronal insult.
Collapse
|
34
|
Garland P, Morton M, Zolnourian A, Durnford A, Gaastra B, Toombs J, Heslegrave AJ, More J, Zetterberg H, Bulters DO, Galea I. Neurofilament light predicts neurological outcome after subarachnoid haemorrhage. Brain 2021; 144:761-768. [PMID: 33517369 PMCID: PMC8041040 DOI: 10.1093/brain/awaa451] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/08/2020] [Accepted: 10/11/2020] [Indexed: 11/17/2022] Open
Abstract
To improve outcome prediction following subarachnoid haemorrhage (SAH), we sought a biomarker integrating early brain injury and multiple secondary pathological processes in a prospective study of 42 non-traumatic SAH patients and 19 control individuals. Neurofilament light (NF-L) was elevated in CSF and serum following SAH. CSF and serum NF-L on Days 1–3 post-SAH strongly predicted modified Rankin score at 6 months, independent of World Federation of Neurosurgical Societies (WFNS) score. NF-L from Day 4 onwards also had a profound impact on outcome. To link NF-L to a SAH-specific pathological process, we investigated NF-L’s relationship with extracellular haemoglobin. Most CSF haemoglobin was not complexed with haptoglobin, yet was able to be bound by exogenous haptoglobin i.e. haemoglobin was scavengeable. CSF scavengeable haemoglobin was strongly predictive of subsequent CSF NF-L. Next, we investigated NF-L efflux from the brain after SAH. Serum and CSF NF-L correlated positively. The serum/CSF NF-L ratio was lower in SAH versus control subjects, in keeping with glymphatic efflux dysfunction after SAH. CSF/serum albumin ratio was increased following SAH versus controls. The serum/CSF NF-L ratio correlated negatively with the CSF/serum albumin ratio, indicating that transfer of the two proteins across the blood–brain interface is dissociated. In summary, NF-L is a strong predictive marker for SAH clinical outcome, adding value to the WFNS score, and is a promising surrogate end point in clinical trials.
Collapse
Affiliation(s)
- Patrick Garland
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Matt Morton
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ardalan Zolnourian
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew Durnford
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ben Gaastra
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jamie Toombs
- UK Dementia Research Institute, University College London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Amanda J Heslegrave
- UK Dementia Research Institute, University College London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - John More
- R&D, Bio Products Laboratory Limited, Elstree, Hertfordshire, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Diederik O Bulters
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
35
|
Warming HK, Schuman EM, Mehta AR. Journey down memory lane. Lancet Neurol 2021; 20:602. [PMID: 34302784 DOI: 10.1016/s1474-4422(21)00217-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
36
|
Buzzi RM, Owczarek CM, Akeret K, Tester A, Pereira N, Butcher R, Brügger-Verdon V, Hardy MP, Illi M, Wassmer A, Vallelian F, Humar R, Hugelshofer M, Buehler PW, Gentinetta T, Schaer DJ. Modular Platform for the Development of Recombinant Hemoglobin Scavenger Biotherapeutics. Mol Pharm 2021; 18:3158-3170. [PMID: 34292741 DOI: 10.1021/acs.molpharmaceut.1c00433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cell-free hemoglobin (Hb) is a driver of disease progression in conditions with intravascular or localized hemolysis. Genetic and acquired anemias or emergency medical conditions such as aneurysmal subarachnoid hemorrhage involve tissue Hb exposure. Haptoglobin (Hp) captures Hb in an irreversible protein complex and prevents its pathophysiological contributions to vascular nitric oxide depletion and tissue oxidation. Preclinical proof-of-concept studies suggest that human plasma-derived Hp is a promising therapeutic candidate for several Hb-driven diseases. Optimizing the efficacy and safety of Hb-targeting biotherapeutics may require structural and functional modifications for specific indications. Improved Hp variants could be designed to achieve the desired tissue distribution, metabolism, and elimination to target hemolytic disease states effectively. However, it is critical to ensure that these modifications maintain the function of Hp. Using transient mammalian gene expression of Hp combined with co-transfection of the pro-haptoglobin processing protease C1r-LP, we established a platform for generating recombinant Hp-variants. We designed an Hpβ-scaffold, which was expressed in this system at high levels as a monomeric unit (mini-Hp) while maintaining the key protective functions of Hp. We then used this Hpβ-scaffold as the basis to develop an initial proof-of-concept Hp fusion protein using human serum albumin as the fusion partner. Next, a hemopexin-Hp fusion protein with bispecific heme and Hb detoxification capacity was generated. Further, we developed a Hb scavenger devoid of CD163 scavenger receptor binding. The functions of these proteins were then characterized for Hb and heme-binding, binding of the Hp-Hb complexes with the clearance receptor CD163, antioxidant properties, and vascular nitric oxide sparing capacity. Our platform is designed to support the generation of innovative Hb scavenger biotherapeutics with novel modes of action and potentially improved formulation characteristics, function, and pharmacokinetics.
Collapse
Affiliation(s)
- Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich 8091, Switzerland
| | | | - Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Zurich 8091, Switzerland
| | - Andrea Tester
- CSL Limited, Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Natasha Pereira
- CSL Limited, Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Rebecca Butcher
- CSL Limited, Bio21 Institute, Parkville, Victoria 3010, Australia
| | | | - Matthew P Hardy
- CSL Limited, Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Marlies Illi
- Research and Development, CSL Behring AG, Bern 3014, Switzerland
| | - Andreas Wassmer
- Research and Development, CSL Behring AG, Bern 3014, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich 8091, Switzerland
| | - Rok Humar
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich 8091, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich, Zurich 8091, Switzerland
| | - Paul W Buehler
- Department of Pathology, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States.,The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | | | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich, Zurich 8091, Switzerland
| |
Collapse
|
37
|
Blevins BL, Vinters HV, Love S, Wilcock DM, Grinberg LT, Schneider JA, Kalaria RN, Katsumata Y, Gold BT, Wang DJJ, Ma SJ, Shade LMP, Fardo DW, Hartz AMS, Jicha GA, Nelson KB, Magaki SD, Schmitt FA, Teylan MA, Ighodaro ET, Phe P, Abner EL, Cykowski MD, Van Eldik LJ, Nelson PT. Brain arteriolosclerosis. Acta Neuropathol 2021; 141:1-24. [PMID: 33098484 PMCID: PMC8503820 DOI: 10.1007/s00401-020-02235-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Brain arteriolosclerosis (B-ASC), characterized by pathologic arteriolar wall thickening, is a common finding at autopsy in aged persons and is associated with cognitive impairment. Hypertension and diabetes are widely recognized as risk factors for B-ASC. Recent research indicates other and more complex risk factors and pathogenetic mechanisms. Here, we describe aspects of the unique architecture of brain arterioles, histomorphologic features of B-ASC, relevant neuroimaging findings, epidemiology and association with aging, established genetic risk factors, and the co-occurrence of B-ASC with other neuropathologic conditions such as Alzheimer's disease and limbic-predominant age-related TDP-43 encephalopathy (LATE). There may also be complex physiologic interactions between metabolic syndrome (e.g., hypertension and inflammation) and brain arteriolar pathology. Although there is no universally applied diagnostic methodology, several classification schemes and neuroimaging techniques are used to diagnose and categorize cerebral small vessel disease pathologies that include B-ASC, microinfarcts, microbleeds, lacunar infarcts, and cerebral amyloid angiopathy (CAA). In clinical-pathologic studies that factored in comorbid diseases, B-ASC was independently associated with impairments of global cognition, episodic memory, working memory, and perceptual speed, and has been linked to autonomic dysfunction and motor symptoms including parkinsonism. We conclude by discussing critical knowledge gaps related to B-ASC and suggest that there are probably subcategories of B-ASC that differ in pathogenesis. Observed in over 80% of autopsied individuals beyond 80 years of age, B-ASC is a complex and under-studied contributor to neurologic disability.
Collapse
Affiliation(s)
- Brittney L Blevins
- Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Seth Love
- University of Bristol and Southmead Hospital, Bristol, BS10 5NB, UK
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Lea T Grinberg
- Department of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- LIM-22, Department of Pathology, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Julie A Schneider
- Departments of Neurology and Pathology, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Samantha J Ma
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Lincoln M P Shade
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University Kentucky, Lexington, KY, 40536, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | | | - Shino D Magaki
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | - Merilee A Teylan
- Department of Epidemiology, University Washington, Seattle, WA, 98105, USA
| | | | - Panhavuth Phe
- Sanders-Brown Center on Aging, University Kentucky, Lexington, KY, 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, Department of Epidemiology, University Kentucky, Lexington, KY, 40536, USA
| | - Matthew D Cykowski
- Departments of Pathology and Genomic Medicine and Neurology, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, Department of Pathology, University of Kentucky, Lexington, KY, 40536, USA.
- Rm 311 Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Avenue, Lexington, KY, 40536, USA.
| |
Collapse
|
38
|
Derry PJ, Vo ATT, Gnanansekaran A, Mitra J, Liopo AV, Hegde ML, Tsai AL, Tour JM, Kent TA. The Chemical Basis of Intracerebral Hemorrhage and Cell Toxicity With Contributions From Eryptosis and Ferroptosis. Front Cell Neurosci 2020; 14:603043. [PMID: 33363457 PMCID: PMC7755086 DOI: 10.3389/fncel.2020.603043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a particularly devastating event both because of the direct injury from space-occupying blood to the sequelae of the brain exposed to free blood components from which it is normally protected. Not surprisingly, the usual metabolic and energy pathways are overwhelmed in this situation. In this review article, we detail the complexity of red blood cell degradation, the contribution of eryptosis leading to hemoglobin breakdown into its constituents, the participants in that process, and the points at which injury can be propagated such as elaboration of toxic radicals through the metabolism of the breakdown products. Two prominent products of this breakdown sequence, hemin, and iron, induce a variety of pathologies including free radical damage and DNA breakage, which appear to include events independent from typical oxidative DNA injury. As a result of this confluence of damaging elements, multiple pathways of injury, cell death, and survival are likely engaged including ferroptosis (which may be the same as oxytosis but viewed from a different perspective) and senescence, suggesting that targeting any single cause will likely not be a sufficient strategy to maximally improve outcome. Combination therapies in addition to safe methods to reduce blood burden should be pursued.
Collapse
Affiliation(s)
- Paul J Derry
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Anh Tran Tram Vo
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Aswini Gnanansekaran
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Joy Mitra
- Department of Neurosurgery, Center for Neuroregeneration, The Houston Methodist Research Institute, Houston, TX, United States
| | - Anton V Liopo
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Muralidhar L Hegde
- Department of Neurosurgery, Center for Neuroregeneration, The Houston Methodist Research Institute, Houston, TX, United States
| | - Ah-Lim Tsai
- Division of Hematology, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - James M Tour
- Department of Chemistry, Rice University, Houston, TX, United States.,Department of Computer Science, George R. Brown School of Engineering, Rice University, Houston, TX, United States.,Department of Materials Science and NanoEngineering, George R. Brown School of Engineering, Rice University, Houston, TX, United States
| | - Thomas A Kent
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States.,Department of Chemistry, Rice University, Houston, TX, United States.,Stanley H. Appel Department of Neurology, Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
39
|
Buehler PW, Humar R, Schaer DJ. Haptoglobin Therapeutics and Compartmentalization of Cell-Free Hemoglobin Toxicity. Trends Mol Med 2020; 26:683-697. [PMID: 32589936 DOI: 10.1016/j.molmed.2020.02.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
Hemolysis and accumulation of cell-free hemoglobin (Hb) in the circulation or in confined tissue compartments such as the subarachnoid space is an important driver of disease. Haptoglobin is the Hb binding and clearance protein in human plasma and an efficient antagonist of Hb toxicity resulting from physiological red blood cell turnover. However, endogenous concentrations of haptoglobin are insufficient to provide protection against Hb-driven disease processes in conditions such as sickle cell anemia, sepsis, transfusion reactions, medical-device associated hemolysis, or after a subarachnoid hemorrhage. As a result, there is increasing interest in developing haptoglobin therapeutics to target 'toxic' cell-free Hb exposures. Here, we discuss key concepts of Hb toxicity and provide a perspective on the use of haptoglobin as a therapeutic protein.
Collapse
Affiliation(s)
- Paul W Buehler
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Rok Humar
- Division of Internal Medicine, University Hospital, Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, University Hospital, Zurich, Switzerland.
| |
Collapse
|