1
|
Hannawi Y. Cerebral Small Vessel Disease: a Review of the Pathophysiological Mechanisms. Transl Stroke Res 2024; 15:1050-1069. [PMID: 37864643 DOI: 10.1007/s12975-023-01195-9] [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: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
Cerebral small vessel disease (cSVD) refers to the age-dependent pathological processes involving the brain small vessels and leading to vascular cognitive impairment, intracerebral hemorrhage, and acute lacunar ischemic stroke. Despite the significant public health burden of cSVD, disease-specific therapeutics remain unavailable due to the incomplete understanding of the underlying pathophysiological mechanisms. Recent advances in neuroimaging acquisition and processing capabilities as well as findings from cSVD animal models have revealed critical roles of several age-dependent processes in cSVD pathogenesis including arterial stiffness, vascular oxidative stress, low-grade systemic inflammation, gut dysbiosis, and increased salt intake. These factors interact to cause a state of endothelial cell dysfunction impairing cerebral blood flow regulation and breaking the blood brain barrier. Neuroinflammation follows resulting in neuronal injury and cSVD clinical manifestations. Impairment of the cerebral waste clearance through the glymphatic system is another potential process that has been recently highlighted contributing to the cognitive decline. This review details these mechanisms and attempts to explain their complex interactions. In addition, the relevant knowledge gaps in cSVD mechanistic understanding are identified and a systematic approach to future translational and early phase clinical research is proposed in order to reveal new cSVD mechanisms and develop disease-specific therapeutics.
Collapse
Affiliation(s)
- Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University, 333 West 10th Ave, Graves Hall 3172C, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Hosoki S, Sachdev PS. Molecular biomarkers for vascular cognitive impairment and dementia: the current status and directions for the future. Neural Regen Res 2024; 19:2579-2580. [PMID: 38808990 PMCID: PMC11168513 DOI: 10.4103/nrr.nrr-d-23-01938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/17/2024] [Accepted: 02/04/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
- Satoshi Hosoki
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
3
|
Qiu J, Peng S, Qu R, Wu L, Xing L, Zhang L, Sun J. New evidence of vascular defects in neurodegenerative diseases revealed by single cell RNA sequencing. Clin Sci (Lond) 2024; 138:1377-1394. [PMID: 39469930 DOI: 10.1042/cs20241658] [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: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Neurodegenerative diseases (NDs) involve the progressive loss of neuronal structure or function in the brain and spinal cord. Despite their diverse etiologies, NDs manifest similar pathologies. Emerging research identifies vascular defects as a previously neglected hallmark of NDs. The development and popularization of single-cell RNA sequencing (scRNA-seq) technologies have significantly advanced our understanding of brain vascular cell types and their molecular characteristics, including gene expression changes at the single-cell level in NDs. These unprecedented insights deepen our understanding of the pathogenic mechanisms underlying NDs. However, the occurrence and role of vascular defects in disease progression remain largely unexplored. In this paper, we systematically summarize recent advances in the structure and organization of the central nervous system vasculature in mice, healthy individuals, and patients with NDs, focussing primarily on disease-specific alterations in vascular cell types or subtypes. Combining scRNA-seq with pathology evidence, we propose that vascular defects, characterized by disruptions in cell types and structural integrity, may serve as common early features of NDs. Finally, we discuss several pathways through which vascular defects in NDs lead to neuronal degeneration. A deeper understanding of the causes and contributions of vascular defects to NDs aids in elucidating the pathogenic mechanisms and developing meaningful therapeutic interventions.
Collapse
Affiliation(s)
- Jiaying Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
4
|
Qi L, Wang C, Deng L, Pan JJ, Suo Q, Wu S, Cai L, Shi X, Sun J, Wang Y, Tang Y, Qiu W, Yang GY, Wang J, Zhang Z. Low-intensity focused ultrasound stimulation promotes stroke recovery via astrocytic HMGB1 and CAMK2N1 in mice. Stroke Vasc Neurol 2024; 9:505-518. [PMID: 38191183 DOI: 10.1136/svn-2023-002614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Low-intensity focused ultrasound stimulation (LIFUS) has been developed to enhance neurological repair and remodelling during the late acute stage of ischaemic stroke in rodents. However, the cellular and molecular mechanisms of neurological repair and remodelling after LIFUS in ischaemic stroke are unclear. METHODS Ultrasound stimulation was treated in adult male mice 7 days after transient middle cerebral artery occlusion. Angiogenesis was measured by laser speckle imaging and histological analyses. Electromyography and fibre photometry records were used for synaptogenesis. Brain atrophy volume and neurobehaviour were assessed 0-14 days after ischaemia. iTRAQ proteomic analysis was performed to explore the differentially expressed protein. scRNA-seq was used for subcluster analysis of astrocytes. Fluorescence in situ hybridisation and Western blot detected the expression of HMGB1 and CAMK2N1. RESULTS Optimal ultrasound stimulation increased cerebral blood flow, and improved neurobehavioural outcomes in ischaemic mice (p<0.05). iTRAQ proteomic analysis revealed that the expression of HMGB1 increased and CAMK2N1 decreased in the ipsilateral hemisphere of the brain at 14 days after focal cerebral ischaemia with ultrasound treatment (p<0.05). scRNA-seq revealed that this expression pattern belonged to a subcluster of astrocytes after LIFUS in the ischaemic brain. LIFUS upregulated HMGB1 expression, accompanied by VEGFA elevation compared with the control group (p<0.05). Inhibition of HMGB1 expression in astrocytes decreased microvessels counts and cerebral blood flow (p<0.05). LIFUS reduced CAMK2N1 expression level, accompanied by increased extracellular calcium ions and glutamatergic synapses (p<0.05). CAMK2N1 overexpression in astrocytes decreased dendritic spines, and aggravated neurobehavioural outcomes (p<0.05). CONCLUSION Our results demonstrated that LIFUS promoted angiogenesis and synaptogenesis after focal cerebral ischaemia by upregulating HMGB1 and downregulating CAMK2N1 in a subcluster of astrocytes, suggesting that LIFUS activated specific astrocyte subcluster could be a key target for ischaemic brain therapy.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Astrocytes/enzymology
- Male
- Recovery of Function
- Disease Models, Animal
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/therapy
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/enzymology
- Mice, Inbred C57BL
- HMGB1 Protein/metabolism
- HMGB1 Protein/genetics
- Neovascularization, Physiologic
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Cerebrovascular Circulation
- Ultrasonic Therapy
- Signal Transduction
- Ischemic Stroke/metabolism
- Ischemic Stroke/physiopathology
- Ischemic Stroke/therapy
- Ischemic Stroke/pathology
- Behavior, Animal
- Time Factors
- Mice
- Proteomics
- Neurogenesis
Collapse
Affiliation(s)
- Lin Qi
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Cheng Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lidong Deng
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Jia-Ji Pan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical Collage, Fudan University, Shanghai, China
| | - Qian Suo
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Shengju Wu
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lin Cai
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xudong Shi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Junfeng Sun
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Yongting Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Weibao Qiu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Jixian Wang
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
5
|
Martineau É, Malescot A, Elmkinssi N, Rungta RL. Distal activity patterns shape the spatial specificity of neurovascular coupling. Nat Neurosci 2024; 27:2101-2114. [PMID: 39232066 DOI: 10.1038/s41593-024-01756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
Neurovascular coupling links brain activity to local changes in blood flow, forming the basis for non-invasive brain mapping. Using multiscale imaging, we investigated how vascular activity spatially relates to neuronal activity elicited by single whiskers across different columns and layers of mouse cortex. Here we show that mesoscopic hemodynamic signals quantitatively reflect neuronal activity across space but are composed of a highly heterogeneous pattern of responses across individual vessel segments that is poorly predicted by local neuronal activity. Rather, this heterogeneity is dependent on vessel directionality, specifically in thalamocortical input layer 4, where capillaries respond preferentially to neuronal activity patterns along their downstream perfusion domain. Thus, capillaries fine-tune blood flow based on distant activity and encode laminar-specific activity patterns. These findings imply that vascular anatomy sets a resolution limit on functional imaging signals, where individual blood vessels inaccurately report neuronal activity in their immediate vicinity but, instead, integrate activity patterns along the vascular arbor.
Collapse
Affiliation(s)
- Éric Martineau
- Centre for Interdisciplinary Research on Brain and Learning (CIRCA), Université de Montréal, Montréal, Quebec, Canada
- Department of Physiology and Pharmacology, Université de Montréal, Montréal, Quebec, Canada
| | - Antoine Malescot
- Centre for Interdisciplinary Research on Brain and Learning (CIRCA), Université de Montréal, Montréal, Quebec, Canada
- Department of Physiology and Pharmacology, Université de Montréal, Montréal, Quebec, Canada
| | - Nouha Elmkinssi
- Centre for Interdisciplinary Research on Brain and Learning (CIRCA), Université de Montréal, Montréal, Quebec, Canada
- Department of Neuroscience, Université de Montréal, Montréal, Quebec, Canada
| | - Ravi L Rungta
- Centre for Interdisciplinary Research on Brain and Learning (CIRCA), Université de Montréal, Montréal, Quebec, Canada.
- Department of Neuroscience, Université de Montréal, Montréal, Quebec, Canada.
- Department of Stomatology, Faculty of Dental Medicine, Université de Montréal, Montréal, Quebec, Canada.
| |
Collapse
|
6
|
Smith EE, Biessels GJ, Gao V, Gottesman RF, Liesz A, Parikh NS, Iadecola C. Systemic determinants of brain health in ageing. Nat Rev Neurol 2024; 20:647-659. [PMID: 39375564 DOI: 10.1038/s41582-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/09/2024]
Abstract
Preservation of brain health is a worldwide priority. The traditional view is that the major threats to the ageing brain lie within the brain itself. Consequently, therapeutic approaches have focused on protecting the brain from these presumably intrinsic pathogenic processes. However, an increasing body of evidence has unveiled a previously under-recognized contribution of peripheral organs to brain dysfunction and damage. Thus, in addition to the well-known impact of diseases of the heart and endocrine glands on the brain, accumulating data suggest that dysfunction of other organs, such as gut, liver, kidney and lung, substantially affects the development and clinical manifestation of age-related brain pathologies. In this Review, a framework is provided to indicate how organ dysfunction can alter brain homeostasis and promote neurodegeneration, with a focus on dementia. We delineate the associations of subclinical dysfunction in specific organs with dementia risk and provide suggestions for public health promotion and clinical management.
Collapse
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Virginia Gao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Neal S Parikh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Liu Y, Liu Q, Shang H, Li J, Chai H, Wang K, Guo Z, Luo T, Liu S, Liu Y, Wang X, Zhang H, Wu C, Song SJ, Yang J. Potential application of natural compounds in ischaemic stroke: Focusing on the mechanisms underlying "lysosomocentric" dysfunction of the autophagy-lysosomal pathway. Pharmacol Ther 2024; 263:108721. [PMID: 39284368 DOI: 10.1016/j.pharmthera.2024.108721] [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: 03/27/2024] [Revised: 07/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Ischaemic stroke (IS) is the second leading cause of death and a major cause of disability worldwide. Currently, the clinical management of IS still depends on restoring blood flow via pharmacological thrombolysis or mechanical thrombectomy, with accompanying disadvantages of narrow therapeutic time window and risk of haemorrhagic transformation. Thus, novel pathophysiological mechanisms and targeted therapeutic candidates are urgently needed. The autophagy-lysosomal pathway (ALP), as a dynamic cellular lysosome-based degradative process, has been comprehensively studied in recent decades, including its upstream regulatory mechanisms and its role in mediating neuronal fate after IS. Importantly, increasing evidence has shown that IS can lead to lysosomal dysfunction, such as lysosomal membrane permeabilization, impaired lysosomal acidity, lysosomal storage disorder, and dysfunctional lysosomal ion homeostasis, which are involved in the IS-mediated defects in ALP function. There is tightly regulated crosstalk between transcription factor EB (TFEB), mammalian target of rapamycin (mTOR) and lysosomal function, but their relationship remains to be systematically summarized. Notably, a growing body of evidence emphasizes the benefits of naturally derived compounds in the treatment of IS via modulation of ALP function. However, little is known about the roles of natural compounds as modulators of lysosomes in the treatment of IS. Therefore, in this context, we provide an overview of the current understanding of the mechanisms underlying IS-mediated ALP dysfunction, from a lysosomal perspective. We also provide an update on the effect of natural compounds on IS, according to their chemical structural types, in different experimental stroke models, cerebral regions and cell types, with a primary focus on lysosomes and autophagy initiation. This review aims to highlight the therapeutic potential of natural compounds that target lysosomal and ALP function for IS treatment.
Collapse
Affiliation(s)
- Yueyang Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hanxiao Shang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jichong Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - He Chai
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kaixuan Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenkun Guo
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tianyu Luo
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shiqi Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yan Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuemei Wang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hangyi Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chunfu Wu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Jingyu Yang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
8
|
Claassen JA. Shifting concepts of autoregulation: Commentary to 'Static autoregulation in humans'. J Cereb Blood Flow Metab 2024; 44:1397-1399. [PMID: 39102511 DOI: 10.1177/0271678x241254676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Advances in imaging techniques have transformed our understanding of cerebral autoregulation. Older imaging techniques provided measurements of cerebral blood flow (CBF) that reflected the average CBF over a window of 10-20 minutes. A key finding, dating back to 1959, was that CBF remained more or less stable over a remarkably wide range of changes in blood pressure. Modern techniques can measure changes in CBF within the time frame of a heartbeat. They have revealed, paradoxically, a remarkable instability of CBF. This commentary attempts to reconcile these seemingly contradictory observations.
Collapse
Affiliation(s)
- Jurgen Ahr Claassen
- Radboud University Medical Center, Department of Geriatrics Nijmegen, the Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
9
|
Anfray A, Schaeffer S, Hattori Y, Santisteban MM, Casey N, Wang G, Strickland M, Zhou P, Holtzman DM, Anrather J, Park L, Iadecola C. A cell-autonomous role for border-associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. Nat Neurosci 2024; 27:2138-2151. [PMID: 39294490 DOI: 10.1038/s41593-024-01757-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Apolipoprotein E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4, we report that border-associated macrophages (BAMs), myeloid cells closely apposed to neocortical microvessels, are both sources and effectors of ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAMs is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell-autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
- Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Samantha Schaeffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Monica M Santisteban
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Michael Strickland
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Moreira TS, Mulkey DK, Takakura AC. Update on vascular control of central chemoreceptors. Exp Physiol 2024; 109:1837-1843. [PMID: 38153366 PMCID: PMC11522829 DOI: 10.1113/ep091329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
At least four mechanisms have been proposed to elucidate how neurons in the retrotrapezoid (RTN) region sense changes in CO2/H+ to regulate breathing (i.e., function as respiratory chemosensors). These mechanisms include: (1) intrinsic neuronal sensitivity to H+ mediated by TASK-2 and GPR4; (2) paracrine activation of RTN neurons by CO2-responsive astrocytes (via a purinergic mechanism); (3) enhanced excitatory synaptic input or disinhibition; and (4) CO2-induced vascular contraction. Although blood flow can influence tissue CO2/H+ levels, there is limited understanding of how control of vascular tone in central CO2 chemosensitive regions might contribute to respiratory output. In this review, we focus on recent evidence that CO2/H+-induced purinergic-dependent vasoconstriction in the ventral parafacial region near RTN neurons supports respiratory chemoreception. This mechanism appears to be unique to the ventral parafacial region and opposite to other brain regions, including medullary chemosensor regions, where CO2/H+ elicits vasodilatation. We speculate that this mechanism helps to maintain CO2/H+ levels in the vicinity of RTN neurons, thereby maintaining the drive to breathe. Important next steps include determining whether disruption of CO2/H+ vascular reactivity contributes to or can be targeted to improve breathing problems in disease states, such as Parkinson's disease.
Collapse
Affiliation(s)
- Thiago S. Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias BiomedicasUniversidade de Sao PauloSao PauloBrazil
| | - Daniel K. Mulkey
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsConnecticutUSA
| | - Ana C. Takakura
- Department of Pharmacology, Instituto de Ciencias BiomedicasUniversidade de Sao PauloSão PauloBrazil
| |
Collapse
|
11
|
Burma JS, Oni IK, Lapointe AP, Rattana S, Schneider KJ, Debert CT, Smirl JD, Dunn JF. Quantifying neurovascular coupling through a concurrent assessment of arterial, capillary, and neuronal activation in humans: A multimodal EEG-fNIRS-TCD investigation. Neuroimage 2024; 302:120910. [PMID: 39486493 DOI: 10.1016/j.neuroimage.2024.120910] [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: 09/12/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND This study explored a novel multimodal neuroimaging approach to assess neurovascular coupling (NVC) in humans using electroencephalography (EEG), functional near-infrared spectroscopy (fNIRS), and transcranial Doppler ultrasound (TCD). METHODS Fifteen participants (nine females; age 19-32) completed concurrent EEG-fNIRS-TCD imaging during motor (finger tapping) and visual ("Where's Waldo?") tasks, with synchronized monitoring of blood pressure, capnography, and heart rate. fNIRS assessed microvascular oxygenation within the frontal, motor, parietal, and occipital cortices, while the middle and posterior cerebral arteries (MCA/PCA) were insonated using TCD. A 16-channel EEG set-up was placed according to the 10-20 system. Wilcoxon signed-rank tests were used to compare physiological responses between the active and resting phases of the tasks, while cross-correlations with zero legs compared cerebral and systemic hemodynamic responses across both tasks. RESULTS Time-frequency analysis demonstrated a reduction in alpha and low beta band power in electrodes C3/C4 during finger tapping (p<0.045) and all electrodes during the Waldo task (all p<0.001). During Waldo, cross-correlation analysis demonstrated the change in oxygenated hemoglobin and cerebral blood velocity had a moderate-to-strong negative correlation with systemic physiological influences, highlighting the measured change resulted from neuronal input. Deoxygenated hemoglobin displayed the greatest negative cross-correlation with the MCA/PCA within the motor cortices and visual during the motor and visual tasks, respectively (range:0.54, -0.82). CONCLUSIONS This investigation demonstrated the feasibility of the proposed EEG-fNIRS-TCD response to comprehensively assess the NVC response within human, specifically quantifying the real-time temporal synchrony between neuronal activation (EEG), microvascular oxygenation changes (fNIRS), and conduit artery velocity alterations (TCD).
Collapse
Affiliation(s)
- Joel S Burma
- Cerebrovascular Concussion Lab, Faculty of Kinesiology, University of Calgary, Alberta, Canada; Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute of Alberta, University of Calgary, Alberta, Canada; Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Integrated Concussion Research Program, University of Calgary, Calgary, Alberta, Canada.
| | - Ibukunoluwa K Oni
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Integrated Concussion Research Program, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Selina Rattana
- Cerebrovascular Concussion Lab, Faculty of Kinesiology, University of Calgary, Alberta, Canada
| | - Kathryn J Schneider
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Integrated Concussion Research Program, University of Calgary, Calgary, Alberta, Canada
| | - Chantel T Debert
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Integrated Concussion Research Program, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan D Smirl
- Cerebrovascular Concussion Lab, Faculty of Kinesiology, University of Calgary, Alberta, Canada; Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute of Alberta, University of Calgary, Alberta, Canada; Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Integrated Concussion Research Program, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F Dunn
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Integrated Concussion Research Program, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Burzyńska M, Woźniak J, Urbański P, Kędziora J, Załuski R, Goździk W, Uryga A. Heart Rate Variability and Cerebral Autoregulation in Patients with Traumatic Brain Injury with Paroxysmal Sympathetic Hyperactivity Syndrome. Neurocrit Care 2024:10.1007/s12028-024-02149-1. [PMID: 39470966 DOI: 10.1007/s12028-024-02149-1] [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: 03/05/2024] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Severe traumatic brain injury (TBI) can lead to transient changes in autonomic nervous system (ANS) functioning and development of paroxysmal sympathetic hyperactivity (PSH) syndrome. Clinical manifestation of ANS disorders may be obscured by therapeutic interventions in TBI. This study aims to analyze ANS metrics and cerebral autoregulation in patients with PSH syndrome to determine their significance in early prognostication. METHODS This single-center retrospective study investigated the relationship between changes in ANS metrics, cerebral autoregulation, and PSH syndrome. Arterial blood pressure and intracranial pressure signals were monitored for 5 days post TBI. ANS metrics included time and frequency domain heart rate variability (HRV) metrics. Cerebral autoregulation was assessed using the pressure reactivity index. RESULTS Sixty-six patients with severe TBI (median age 33 [interquartile range 26-50] years) were analyzed, and PSH was confirmed in nine cases. Impairment of cerebral autoregulation was observed in 67% of patients with PSH and 72% without the syndrome. Patients with PSH had higher HRV in the low-frequency range (LF; 253 ± 178 vs. 176 ± 227 ms2; p = 0.035) and lower heart rates (HRs; 70 ± 7 vs. 78 ± 19 bpm; p = 0.027) compared to those without PSH. A receiver operating characteristic curve analysis indicated that HR (area under the curve (AUC) = 0.73, p = 0.006) and HRV in the LF (AUC = 0.70, p = 0.009) are moderate predictors of PSH. In the multiple logistic regression model for PSH, diffuse axonal trauma (odds ratio (OR) = 10.82, 95% confidence interval (CI) = 1.70-68.98, p = 0.012) and HR (OR = 0.91, 95% CI 0.84-0.98, p = 0.021) were significant factors. CONCLUSIONS Elevated HRV in the LF and decreased HR may serve as early predictors of PSH syndrome development, particularly in patients with diffuse axonal trauma. Further research is needed to investigate the utility of the cerebral autoregulation-ANS relationship in PSH prognostication.
Collapse
Affiliation(s)
- Małgorzata Burzyńska
- Clinical Department of Anesthesiology and Intensive Care, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Jowita Woźniak
- Department of Neurosurgery, Wroclaw University Hospital, Wroclaw, Poland
| | - Piotr Urbański
- Clinical Department of Anesthesiology and Intensive Care, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Jarosław Kędziora
- Clinical Department of Anesthesiology and Intensive Care, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Rafał Załuski
- Clinical Department of Neurosurgery, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Waldemar Goździk
- Clinical Department of Anesthesiology and Intensive Care, Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Agnieszka Uryga
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370, Wrocław, Poland.
| |
Collapse
|
13
|
Dar IA, Khan IR, Johnson TW, Helmy SM, Cardona JI, Escobar S, Selioutski O, Marinescu MA, Zhang CT, Proctor AR, AbdAllah N, Busch DR, Maddox RK, Choe R. Wavelet and time-based cerebral autoregulation analysis using diffuse correlation spectroscopy on adults undergoing extracorporeal membrane oxygenation therapy. PLoS One 2024; 19:e0299752. [PMID: 39471182 PMCID: PMC11521301 DOI: 10.1371/journal.pone.0299752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/23/2024] [Indexed: 11/01/2024] Open
Abstract
INTRODUCTION Adult patients who have suffered acute cardiac or pulmonary failure are increasingly being treated using extracorporeal membrane oxygenation (ECMO), a cardiopulmonary bypass technique. While ECMO has improved the long-term outcomes of these patients, neurological injuries can occur from underlying illness or ECMO itself. Cerebral autoregulation (CA) allows the brain to maintain steady perfusion during changes in systemic blood pressure. Dysfunctional CA is a marker of acute brain injury and can worsen neurologic damage. Monitoring CA using invasive modalities can be risky in ECMO patients due to the necessity of anticoagulation therapy. Diffuse correlation spectroscopy (DCS) measures cerebral blood flow continuously, noninvasively, at the bedside, and can monitor CA. In this study, we compare DCS-based markers of CA in veno-arterial ECMO patients with and without acute brain injury. METHODS Adults undergoing ECMO were prospectively enrolled at a single tertiary hospital and underwent DCS and arterial blood pressure monitoring during ECMO. Neurologic injuries were identified using brain computerized tomography (CT) scans obtained in all patients. CA was calculated over a twenty-minute window via wavelet coherence analysis (WCA) over 0.05 Hz to 0.1 Hz and a Pearson correlation (DCSx) between cerebral blood flow measured by DCS and mean arterial pressure. RESULTS Eleven ECMO patients who received CT neuroimaging were recruited. 5 (45%) patients were found to have neurologic injury. CA indices WCOH, the area under the curve of the WCA, were significantly higher for patients with neurological injuries compared to those without neurological injuries (right hemisphere p = 0.041, left hemisphere p = 0.041). %DCSx, percentage of time DCSx was above a threshold 0.4, were not significantly higher (right hemisphere p = 0.268, left hemisphere p = 0.073). CONCLUSION DCS can be used to detect differences in CA for ECMO patients with neurological injuries compared to uninjured patients using WCA.
Collapse
Affiliation(s)
- Irfaan A. Dar
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, United States of America
| | - Imad R. Khan
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Thomas W. Johnson
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Samantha Marie Helmy
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Jeronimo I. Cardona
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Samantha Escobar
- Clinical and Translational Sciences Program, University of Rochester, Rochester, New York, United States of America
| | - Olga Selioutski
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, United States of America
- Department of Neurology, University of Mississippi, Jackson, Mississippi, United States of America
| | - Mark A. Marinescu
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Chloe T. Zhang
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, United States of America
| | - Ashley R. Proctor
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Noura AbdAllah
- Department of Biology, University of Rochester, Rochester, New York, United States of America
| | - David R. Busch
- Departments of Anesthesiology and Pain Management, Neurology and Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ross K. Maddox
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, United States of America
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Regine Choe
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, United States of America
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, New York, United States of America
| |
Collapse
|
14
|
Mendiola PJ, O'Herron P, Xie K, Brands MW, Bush W, Patterson RE, Di Stefano V, Filosa JA. Blood pressure variability compromises vascular function in middle-aged mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619509. [PMID: 39484398 PMCID: PMC11526967 DOI: 10.1101/2024.10.21.619509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Blood pressure variability (BPV) has emerged as a novel risk factor for cognitive decline and dementia, independent of alterations in average blood pressure (BP). However, the underlying consequences of large BP fluctuations on the neurovascular complex are unknown. We developed a novel mouse model of BPV in middle-aged mice based on intermittent Angiotensin II infusions. Using radio telemetry, we demonstrated that the 24-hr BP averages of these mice were similar to controls, indicating BPV without hypertension. Chronic (20-25 days) BPV led to a blunted bradycardic response and cognitive deficits. Two-photon imaging of parenchymal arterioles showed enhanced pressure-evoked constrictions (myogenic response) in BPV mice. Sensory stimulus-evoked dilations (neurovascular coupling) were greater at higher BP levels in control mice, but this pressure-dependence was lost in BPV mice. Our findings support the notion that large BP variations impair vascular function at the neurovascular complex and contribute to cognitive decline.
Collapse
|
15
|
Lacoste B. Sensors in the microvascular web: Vital but vulnerable. Proc Natl Acad Sci U S A 2024; 121:e2417137121. [PMID: 39401368 PMCID: PMC11513975 DOI: 10.1073/pnas.2417137121] [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: 10/30/2024] Open
Affiliation(s)
- Baptiste Lacoste
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, ONK1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ONK1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ONK1H 8M5, Canada
| |
Collapse
|
16
|
Gal Z, Torok D, Gonda X, Eszlari N, Anderson IM, Deakin B, Petschner P, Juhasz G, Bagdy G. New Evidence for the Role of the Blood-Brain Barrier and Inflammation in Stress-Associated Depression: A Gene-Environment Analysis Covering 19,296 Genes in 109,360 Humans. Int J Mol Sci 2024; 25:11332. [PMID: 39457114 PMCID: PMC11508422 DOI: 10.3390/ijms252011332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Mounting evidence supports the key role of the disrupted integrity of the blood-brain barrier (BBB) in stress- and inflammation-associated depression. We assumed that variations in genes regulating the expression and coding proteins constructing and maintaining this barrier, along with those involved in inflammation, have a predisposing or protecting role in the development of depressive symptoms after experiencing severe stress. To prove this, genome-by-environment (GxE) interaction analyses were conducted on 6.26 M SNPS covering 19,296 genes on PHQ9 depression in interaction with adult traumatic events scores in the UK Biobank (n = 109,360) in a hypothesis-free setup. Among the 63 genes that were significant in stress-connected depression, 17 were associated with BBB, 23 with inflammatory processes, and 4 with neuroticism. Compared to all genes, the enrichment of significant BBB-associated hits was 3.82, and those of inflammation-associated hits were 1.59. Besides some sex differences, CSMD1 and PTPRD, encoding proteins taking part in BBB integrity, were the most significant hits in both males and females. In conclusion, the identified risk genes and their encoded proteins could provide biomarkers or new drug targets to promote BBB integrity and thus prevent or decrease stress- and inflammation-associated depressive symptoms, and possibly infection, e.g., COVID-19-associated mental and neurological symptoms.
Collapse
Affiliation(s)
- Zsofia Gal
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Dora Torok
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Xenia Gonda
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
- Department of Psychiatry and Psychotherapy, Semmelweis University, 1083 Budapest, Hungary
| | - Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Ian Muir Anderson
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9NT, UK; (I.M.A.); (B.D.)
| | - Bill Deakin
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9NT, UK; (I.M.A.); (B.D.)
| | - Peter Petschner
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
- Bioinformatics Center, Institute of Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Realization of Sustainable Society, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| |
Collapse
|
17
|
Silvestro M, Esposito F, De Rosa AP, Orologio I, Trojsi F, Tartaglione L, García-Polo P, Tedeschi G, Tessitore A, Cirillo M, Russo A. Reduced neurovascular coupling of the visual network in migraine patients with aura as revealed with arterial spin labeling MRI: is there a demand-supply mismatch behind the scenes? J Headache Pain 2024; 25:180. [PMID: 39407094 PMCID: PMC11481770 DOI: 10.1186/s10194-024-01885-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Although neuroimaging investigations have consistently demonstrated that "hyperresponsive" and "hyperconnected" visual cortices may represent the functional substrate of cortical spreading depolarization in patients with migraine with aura, the mechanisms which underpin the brain "tendency" to ignite the cortical spreading depolarization and, consequently, aura phenomenon are still matter of debate. Considering that triggers able to induce aura phenomenon constrain brain to increase global (such as physical activity, stressors and sleep abnormalities) or local (such as bright light visual stimulations) energy demand, a vascular supply unable to satisfy the increased energy requirement could be hypothesized in these patients. METHODS Twenty-three patients with migraine with aura, 25 patients with migraine without aura and 20 healthy controls underwent a 3-Tesla MRI study. Cerebral blood flow and local functional connectivity (regional homogeneity) maps were obtained and registered to the MNI space where 100 cortical regions were derived using a functional local-global normative parcellation. A surrogate estimate of the regional neurovascular coupling for each subject was obtained at each parcel from the correlation coefficient between the z-scored ReHo map and the z-scored cerebral blood flow maps. RESULTS A significantly higher regional cerebral blood flow across the visual cortex of both hemispheres (i.e. fusiform and lingual gyri) was detected in migraine with aura patients when compared to patients with migraine without aura (p < 0.05, corrected for multiple comparisons). Concomitantly, a significantly reduced neurovascular coupling (p < 0.05, false discovery rate corrected) in the primary visual cortex parcel (VIS-4) of the large-scale visual network was observed in the left hemisphere of patients with migraine with aura (0.23±0.03), compared to both patients with migraine without aura (0.32±0.05) and healthy controls (0.29±0.05). CONCLUSIONS Visual cortex neurovascular "decoupling" might represent the "link" between the exposure to trigger factors and aura phenomenon ignition. While physiological vascular oversupply may compensate neurovascular demand-supply at rest, it becomes inadequate in case of increased energy demand (e.g. when patients face with trigger factors) paving the way to the aura phenomenon ignition in patients with migraine with aura. Whether preventive treatments may exert their therapeutic activity on migraine with aura restoring the energy demands and cerebral blood flow trade-off within the visual network should be further investigated.
Collapse
Affiliation(s)
- Marcello Silvestro
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fabrizio Esposito
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandro Pasquale De Rosa
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ilaria Orologio
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesca Trojsi
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lorenzo Tartaglione
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Gioacchino Tedeschi
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandro Tessitore
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Cirillo
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Russo
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
18
|
Lim XR, Abd-Alhaseeb MM, Ippolito M, Koide M, Senatore AJ, Plante C, Hariharan A, Weir N, Longden TA, Laprade KA, Stafford JM, Ziemens D, Schwaninger M, Wenzel J, Postnov DD, Harraz OF. Endothelial Piezo1 channel mediates mechano-feedback control of brain blood flow. Nat Commun 2024; 15:8686. [PMID: 39375369 PMCID: PMC11458797 DOI: 10.1038/s41467-024-52969-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
Hyperemia in response to neural activity is essential for brain health. A hyperemic response delivers O2 and nutrients, clears metabolic waste, and concomitantly exposes cerebrovascular endothelial cells to hemodynamic forces. While neurovascular research has primarily centered on the front end of hyperemia-neuronal activity-to-vascular response-the mechanical consequences of hyperemia have gone largely unexplored. Piezo1 is an endothelial mechanosensor that senses hyperemia-associated forces. Using genetic mouse models and pharmacologic approaches to manipulate endothelial Piezo1 function, we evaluated its role in blood flow control and whether it impacts cognition. We provide evidence of a built-in brake system that sculpts hyperemia, and specifically show that Piezo1 activation triggers a mechano-feedback system that promotes blood flow recovery to baseline. Further, genetic Piezo1 modification led to deficits in complementary memory tasks. Collectively, our findings establish a role for endothelial Piezo1 in cerebral blood flow regulation and a role in its behavioral sequelae.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Michael Ippolito
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Masayo Koide
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Amanda J Senatore
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Curtis Plante
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Kathryn A Laprade
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - James M Stafford
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Dorothea Ziemens
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Jan Wenzel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Dmitry D Postnov
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, 8200, Denmark
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
19
|
Banks WA, Rhea EM, Reed MJ, Erickson MA. The penetration of therapeutics across the blood-brain barrier: Classic case studies and clinical implications. Cell Rep Med 2024:101760. [PMID: 39383873 DOI: 10.1016/j.xcrm.2024.101760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/20/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024]
Abstract
The blood-brain barrier (BBB) plays central roles in the maintenance and health of the brain. Its mechanisms to safeguard the brain against xenobiotics and endogenous toxins also make the BBB the primary obstacle to the development of drugs for the central nervous system (CNS). Here, we review classic examples of the intersection of clinical medicine, drug delivery, and the BBB. We highlight the role of lipid solubility (heroin), saturable brain-to-blood (efflux: opiates) and blood-to-brain (influx: nutrients, vitamins, and minerals) transport systems, and adsorptive transcytosis (viruses and incretin receptor agonists). We examine how the disruption of the BBB that occurs in certain diseases (tumors) can also be modulated (osmotic agents and microbubbles) and used to deliver treatments, and the role of extracellular pathways in gaining access to the CNS (albumin and antibodies). In summary, this review provides a historical perspective of the key role of the BBB in delivery of drugs to the brain in health and disease.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA.
| | - Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - May J Reed
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| |
Collapse
|
20
|
Kartal A, Robba C, Helmy A, Wolf S, Aries MJH. How to Define and Meet Blood Pressure Targets After Traumatic Brain Injury: A Narrative Review. Neurocrit Care 2024; 41:369-385. [PMID: 38982005 PMCID: PMC11377672 DOI: 10.1007/s12028-024-02048-5] [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: 02/14/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) poses a significant challenge to healthcare providers, necessitating meticulous management of hemodynamic parameters to optimize patient outcomes. This article delves into the critical task of defining and meeting continuous arterial blood pressure (ABP) and cerebral perfusion pressure (CPP) targets in the context of severe TBI in neurocritical care settings. METHODS We narratively reviewed existing literature, clinical guidelines, and emerging technologies to propose a comprehensive approach that integrates real-time monitoring, individualized cerebral perfusion target setting, and dynamic interventions. RESULTS Our findings emphasize the need for personalized hemodynamic management, considering the heterogeneity of patients with TBI and the evolving nature of their condition. We describe the latest advancements in monitoring technologies, such as autoregulation-guided ABP/CPP treatment, which enable a more nuanced understanding of cerebral perfusion dynamics. By incorporating these tools into a proactive monitoring strategy, clinicians can tailor interventions to optimize ABP/CPP and mitigate secondary brain injury. DISCUSSION Challenges in this field include the lack of standardized protocols for interpreting multimodal neuromonitoring data, potential variability in clinical decision-making, understanding the role of cardiac output, and the need for specialized expertise and customized software to have individualized ABP/CPP targets regularly available. The patient outcome benefit of monitoring-guided ABP/CPP target definitions still needs to be proven in patients with TBI. CONCLUSIONS We recommend that the TBI community take proactive steps to translate the potential benefits of personalized ABP/CPP targets, which have been implemented in certain centers, into a standardized and clinically validated reality through randomized controlled trials.
Collapse
Affiliation(s)
- Ahmet Kartal
- University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany.
| | - Chiara Robba
- Anesthesia and Intensive Care, IRCCS Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Sciences, University of Genoa, Genoa, Italy
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Stefan Wolf
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel J H Aries
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht University, Maastricht, The Netherlands
- Institute of Mental Health and Neurosciences, University Maastricht, Maastricht, The Netherlands
| |
Collapse
|
21
|
van der Horn HJ, Vakhtin AA, Julio K, Nitschke S, Shaff N, Dodd AB, Erhardt E, Phillips JP, Pirio Richardson S, Deligtisch A, Stewart M, Suarez Cedeno G, Meles SK, Mayer AR, Ryman SG. Parkinson's disease cerebrovascular reactivity pattern: A feasibility study. J Cereb Blood Flow Metab 2024; 44:1774-1786. [PMID: 38578669 PMCID: PMC11494834 DOI: 10.1177/0271678x241241895] [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: 10/06/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
A mounting body of research points to cerebrovascular dysfunction as a fundamental element in the pathophysiology of Parkinson's disease (PD). In the current feasibility study, blood-oxygen-level-dependent (BOLD) MRI was used to measure cerebrovascular reactivity (CVR) in response to hypercapnia in 26 PD patients and 16 healthy controls (HC), and aimed to find a multivariate pattern specific to PD. Whole-brain maps of CVR amplitude (i.e., magnitude of response to CO2) and latency (i.e., time to reach maximum amplitude) were computed, which were further analyzed using scaled sub-profile model principal component analysis (SSM-PCA) with leave-one-out cross-validation. A meaningful pattern based on CVR latency was identified, which was named the PD CVR pattern (PD-CVRP). This pattern was characterized by relatively increased latency in basal ganglia, sensorimotor cortex, supplementary motor area, thalamus and visual cortex, as well as decreased latency in the cerebral white matter, relative to HC. There were no significant associations with clinical measures, though sample size may have limited our ability to detect significant associations. In summary, the PD-CVRP highlights the importance of cerebrovascular dysfunction in PD, and may be a potential biomarker for future clinical research and practice.
Collapse
Affiliation(s)
- Harm Jan van der Horn
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Andrei A Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Kayla Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Stephanie Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Nicholas Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Andrew B Dodd
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Erik Erhardt
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, USA
| | - John P Phillips
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Sarah Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
- New Mexico VA Health Care System, Albuquerque, NM, USA
| | - Amanda Deligtisch
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Melanie Stewart
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Gerson Suarez Cedeno
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Sanne K Meles
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrew R Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
| | - Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, NM, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
22
|
Yu X, Xu D, Hu J, Yu Y, Wang L, Jiang B, Zhang M. Exploring the Impact of Hemoglobin on Cerebral Blood Flow in Arterial Territories and Surgical Outcomes: Potential Implications for Moyamoya Disease Treatment. J Am Heart Assoc 2024; 13:e035387. [PMID: 39344645 DOI: 10.1161/jaha.124.035387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/01/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Changes in levels of hemoglobin would result in alterations of cerebral blood flow (CBF). However, the impact of hemoglobin on CBF in moyamoya disease (MMD) remains largely unknown. This study sought to determine whether CBF would be influenced by hemoglobin before surgical revascularization and to analyze the relationships between hemoglobin and CBF with clinical outcome after surgery in patients with MMD. METHODS AND RESULTS We prospectively enrolled adult patients with MMD undergoing surgical revascularization between June 2020 and December 2022. Preoperative CBF was measured in the territories of anterior, middle, and posterior cerebral arteries (ACA, MCA, and PCA, respectively) using 3-dimensional pseudo-continuous arterial spin labeling magnetic resonance imaging. Clinical outcome at 1 year after surgery was evaluated using the modified Rankin Scale. A total of 60 patients with MMD were included, with 25% (n=15) experiencing unfavorable outcomes. Patients with MMD exhibited lower CBF (ACA: P=0.007; MCA: P<0.001; PCA: P=0.014), compared with healthy controls (n=40). Hemoglobin was negatively and significantly associated with CBF (ACA: β=-0.45, P<0.001; MCA: β=-0.38, P<0.001; PCA: β=-0.54, P<0.001). CBF rather than hemoglobin was significantly related with clinical outcome (ACA: P<0.001; MCA: P<0.001; PCA: P=0.001), and CBF showed high discrimination in predicting clinical outcome (ACA: area under the curve, 0.84; MCA: area under the curve, 0.84; PCA: area under the curve, 0.80). CONCLUSIONS Our findings demonstrate that hemoglobin significantly influences CBF, and CBF has a high predictive value for clinical outcome in MMD. The optimal hemoglobin level before surgical revascularization should be further investigated.
Collapse
Affiliation(s)
- Xinfeng Yu
- Department of Radiology The Second Affiliated Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Duo Xu
- Department of Radiology The Second Affiliated Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Junwen Hu
- Department of Neurosurgery The Second Affiliated Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Yannan Yu
- Department of Radiology University of California San Francisco San Francisco CA
| | - Lin Wang
- Department of Neurosurgery The Second Affiliated Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Biao Jiang
- Department of Radiology The Second Affiliated Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Minming Zhang
- Department of Radiology The Second Affiliated Hospital, Zhejiang University School of Medicine Hangzhou China
| |
Collapse
|
23
|
Shadman J, Panahpour H, Alipour MR, Salimi A, Shahabi P, Azar SS. Investigating the therapeutic effects of nimodipine on vasogenic cerebral edema and blood-brain barrier impairment in an ischemic stroke rat model. Neuropharmacology 2024; 257:110054. [PMID: 38950691 DOI: 10.1016/j.neuropharm.2024.110054] [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: 03/24/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Vasogenic brain edema, a potentially life-threatening consequence following an acute ischemic stroke, is a major clinical problem. This research aims to explore the therapeutic benefits of nimodipine, a calcium channel blocker, in mitigating vasogenic cerebral edema and preserving blood-brain barrier (BBB) function in an ischemic stroke rat model. In this research, animals underwent the induction of ischemic stroke via a 60-min blockage of the middle cerebral artery and treated with a nonhypotensive dose of nimodipine (1 mg/kg/day) for a duration of five days. The wet/dry method was employed to identify cerebral edema, and the Evans blue dye extravasation technique was used to assess the permeability of the BBB. Furthermore, immunofluorescence staining was utilized to assess the protein expression levels of matrix metalloproteinase-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1). The study also examined mitochondrial function by evaluating mitochondrial swelling, succinate dehydrogenase (SDH) activity, the collapse of mitochondrial membrane potential (MMP), and the generation of reactive oxygen species (ROS). Post-stroke administration of nimodipine led to a significant decrease in cerebral edema and maintained the integrity of the BBB. The protective effects observed were associated with a reduction in cell apoptosis as well as decreased expression of MMP-9 and ICAM-1. Furthermore, nimodipine was observed to reduce mitochondrial swelling and ROS levels while simultaneously restoring MMP and SDH activity. These results suggest that nimodipine may reduce cerebral edema and BBB breakdown caused by ischemia/reperfusion. This effect is potentially mediated through the reduction of MMP-9 and ICAM-1 levels and the enhancement of mitochondrial function.
Collapse
Affiliation(s)
- Javad Shadman
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamdollah Panahpour
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | | | - Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saied Salimpour Azar
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
24
|
O’Grady BJ, McCall AS, Cullison S, Chavarria D, Kjar A, Schrag MS, Lippmann ES. Anatomically and Physiologically Accurate Engineered Neurovascular Unit and Blood-Brain Barrier Model Using Microvessels Isolated from Postmortem Human Brain Tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615283. [PMID: 39386654 PMCID: PMC11463414 DOI: 10.1101/2024.09.26.615283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Brain vasculature is a complex and heterogeneous physiological structure that serves specialized roles in maintaining brain health and homeostasis. There is substantial interest in developing representative human models of the brain vasculature for drug screening and disease modeling applications. Many contemporary strategies have focused on culturing neurovascular cell types in hydrogels and microdevices, but it remains challenging to achieve anatomically relevant vascular structures that have physiologically similar function to their in vivo counterparts. Here, we present a strategy for isolating microvessels from cryopreserved human cortical tissue and culturing these vessels in a biomimetic gelatin-based hydrogel contained in a microfluidic device. We provide histological evidence of arteriole and capillary architectures within hydrogels, as well as anastomosis to the hydrogel edges allowing lumen perfusion. In capillaries, we demonstrate restricted diffusion of a 10 kDa dextran, indicating intact passive blood-brain barrier function. We anticipate this bona fide human brain vasculature-on-a-chip will be useful for various biotechnology applications.
Collapse
Affiliation(s)
- Brian J. O’Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - A. Scott McCall
- Department of Pulmonary and Critical Care, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel Cullison
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Daniel Chavarria
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Matthew S. Schrag
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S. Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Pulmonary and Critical Care, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
25
|
Xie Y, Yang F, He L, Huang H, Chao M, Cao H, Hu Y, Fan Z, Zhai Y, Zhao W, Liu X, Zhao R, Xiao B, Shi X, Luo Y, Yin J, Feng D, Hugnot JP, Muhl L, Dimberg A, Betsholtz C, Zhang Y, Wang L, Zhang L. Single-cell dissection of the human blood-brain barrier and glioma blood-tumor barrier. Neuron 2024; 112:3089-3105.e7. [PMID: 39191260 DOI: 10.1016/j.neuron.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/01/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024]
Abstract
The blood-brain barrier (BBB) serves as a crucial vascular specialization, shielding and nourishing brain neurons and glia while impeding drug delivery. Here, we conducted single-cell mRNA sequencing of human cerebrovascular cells from 13 surgically resected glioma samples and adjacent normal brain tissue. The transcriptomes of 103,230 cells were mapped, including 57,324 endothelial cells (ECs) and 27,703 mural cells (MCs). Both EC and MC transcriptomes originating from lower-grade glioma were indistinguishable from those of normal brain tissue, whereas transcriptomes from glioblastoma (GBM) displayed a range of abnormalities. Among these, we identified LOXL2-dependent collagen modification as a common GBM-dependent trait and demonstrated that inhibiting LOXL2 enhanced chemotherapy efficacy in both murine and human patient-derived xenograft (PDX) GBM models. Our comprehensive single-cell RNA sequencing-based molecular atlas of the human BBB, coupled with insights into its perturbations in GBM, holds promise for guiding future investigations into brain health, pathology, and therapeutic strategies.
Collapse
Affiliation(s)
- Yuan Xie
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Fan Yang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China
| | - Haiyan Cao
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China
| | - Yaqin Hu
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China
| | - Zhicheng Fan
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China
| | - Yaohong Zhai
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Wenjian Zhao
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China
| | - Xian Liu
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Ruozhu Zhao
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Bing Xiao
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xinxin Shi
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yuancheng Luo
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Jinlong Yin
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China
| | - Jean-Philippe Hugnot
- Jinfeng Laboratory, Chongqing 401329, China; Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Lars Muhl
- Department of Medicine Huddinge, Karolinska Institute, 14157 Huddinge, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185 Uppsala, Sweden; Department of Medicine Huddinge, Karolinska Institute, 14157 Huddinge, Sweden.
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi'an 710038, China.
| | - Lei Zhang
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
26
|
Marchant JK, Ferris NG, Grass D, Allen MS, Gopalakrishnan V, Olchanyi M, Sehgal D, Sheft M, Strom A, Bilgic B, Edlow B, Hillman EMC, Juttukonda MR, Lewis L, Nasr S, Nummenmaa A, Polimeni JR, Tootell RBH, Wald LL, Wang H, Yendiki A, Huang SY, Rosen BR, Gollub RL. Mesoscale Brain Mapping: Bridging Scales and Modalities in Neuroimaging - A Symposium Review. Neuroinformatics 2024:10.1007/s12021-024-09686-2. [PMID: 39312131 DOI: 10.1007/s12021-024-09686-2] [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] [Accepted: 08/20/2024] [Indexed: 10/20/2024]
Abstract
Advances in the spatiotemporal resolution and field-of-view of neuroimaging tools are driving mesoscale studies for translational neuroscience. On October 10, 2023, the Center for Mesoscale Mapping (CMM) at the Massachusetts General Hospital (MGH) Athinoula A. Martinos Center for Biomedical Imaging and the Massachusetts Institute of Technology (MIT) Health Sciences Technology based Neuroimaging Training Program (NTP) hosted a symposium exploring the state-of-the-art in this rapidly growing area of research. "Mesoscale Brain Mapping: Bridging Scales and Modalities in Neuroimaging" brought together researchers who use a broad range of imaging techniques to study brain structure and function at the convergence of the microscopic and macroscopic scales. The day-long event centered on areas in which the CMM has established expertise, including the development of emerging technologies and their application to clinical translational needs and basic neuroscience questions. The in-person symposium welcomed more than 150 attendees, including 57 faculty members, 61 postdoctoral fellows, 35 students, and four industry professionals, who represented institutions at the local, regional, and international levels. The symposium also served the training goals of both the CMM and the NTP. The event content, organization, and format were planned collaboratively by the faculty and trainees. Many CMM faculty presented or participated in a panel discussion, thus contributing to the dissemination of both the technologies they have developed under the auspices of the CMM and the findings they have obtained using those technologies. NTP trainees who benefited from the symposium included those who helped to organize the symposium and/or presented posters and gave "flash" oral presentations. In addition to gaining experience from presenting their work, they had opportunities throughout the day to engage in one-on-one discussions with visiting scientists and other faculty, potentially opening the door to future collaborations. The symposium presentations provided a deep exploration of the many technological advances enabling progress in structural and functional mesoscale brain imaging. Finally, students worked closely with the presenting faculty to develop this report summarizing the content of the symposium and putting it in the broader context of the current state of the field to share with the scientific community. We note that the references cited here include conference abstracts corresponding to the symposium poster presentations.
Collapse
Affiliation(s)
- Joshua K Marchant
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA.
| | - Natalie G Ferris
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.
- Harvard Biophysics Graduate Program, Cambridge, MA, USA.
| | - Diana Grass
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Magdelena S Allen
- Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Vivek Gopalakrishnan
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mark Olchanyi
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Devang Sehgal
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Maxina Sheft
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Amelia Strom
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Berkin Bilgic
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Brian Edlow
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth M C Hillman
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Radiology, Columbia University, New York, NY, USA
| | - Meher R Juttukonda
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Laura Lewis
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shahin Nasr
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aapo Nummenmaa
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jonathan R Polimeni
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Roger B H Tootell
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lawrence L Wald
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Hui Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Anastasia Yendiki
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Bruce R Rosen
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Randy L Gollub
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Bassi I, Grunspan M, Hen G, Ravichandran KA, Moshe N, Gutierrez-Miranda L, Safriel SR, Kostina D, Shen A, Ruiz de Almodovar C, Yaniv K. Endolysosomal dysfunction in radial glia progenitor cells leads to defective cerebral angiogenesis and compromised blood-brain barrier integrity. Nat Commun 2024; 15:8158. [PMID: 39289367 PMCID: PMC11408700 DOI: 10.1038/s41467-024-52365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
The neurovascular unit (NVU) is a complex multicellular structure that helps maintain cerebral homeostasis and blood-brain barrier (BBB) integrity. While extensive evidence links NVU alterations to cerebrovascular diseases and neurodegeneration, the underlying molecular mechanisms remain unclear. Here, we use zebrafish embryos carrying a mutation in Scavenger Receptor B2, a highly conserved endolysosomal protein expressed predominantly in Radial Glia Cells (RGCs), to investigate the interplay among different NVU components. Through live imaging and genetic manipulations, we demonstrate that compromised acidification of the endolysosomal compartment in mutant RGCs leads to impaired Notch3 signaling, thereby inducing excessive neurogenesis and reduced glial differentiation. We further demonstrate that alterations to the neuron/glia balance result in impaired VEGF and Wnt signaling, leading to severe vascular defects, hemorrhages, and a leaky BBB. Altogether, our findings provide insights into NVU formation and function and offer avenues for investigating diseases involving white matter defects and vascular abnormalities.
Collapse
Affiliation(s)
- Ivan Bassi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Grunspan
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gideon Hen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kishore A Ravichandran
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Noga Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Laura Gutierrez-Miranda
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav R Safriel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Kostina
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Amitay Shen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Carmen Ruiz de Almodovar
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Bonn, Germany
| | - Karina Yaniv
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
28
|
Wu ZY, Luo YF, Lu YP, Zhang YX, Han F. Targeting Brain Endothelial Gasdermin D: A Shortcut to Remodel the Blood-Brain Barrier. Neurosci Bull 2024:10.1007/s12264-024-01300-4. [PMID: 39276179 DOI: 10.1007/s12264-024-01300-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/20/2024] [Indexed: 09/16/2024] Open
Affiliation(s)
- Zhou-Yue Wu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yi-Fan Luo
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ya-Ping Lu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China.
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
29
|
Jin Y, Zhou X, Chen L, Xu X, Yan W, Wang Q, Lin Y, Ding X. Framework Nucleic Acids Loaded with Quercetin: Protecting Retinal Neurovascular Unit via the Protein Kinase B/Heme Oxygenase-1 Pathway. ACS NANO 2024. [PMID: 39268926 DOI: 10.1021/acsnano.4c05845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Retinal neovascular disease is a leading cause of vision loss and blindness globally. It occurs when abnormal new blood vessels form in the retina. In this study, we utilized tetrahedral framework nucleic acids (tFNAs) as vehicles to load quercetin (QUE), a small-molecule flavonoid, forming a deoxyribonucleic acid (DNA) nanocomplex, tFNAs-QUE. Our data show this nanocomplex inhibits pathological neovascularization, reduces the area of retinal nonperfusion area, protects retinal neurons, and preserves the visual function. Further, we discovered that tFNAs-QUE selectively upregulates the AKT/Nrf2/HO-1 signaling pathway, which can suppress pathological vascular growth and exert antioxidative effects. Therefore, this study presents a promising small-molecule-loading mechanism for the treatment of ischemic retinal diseases.
Collapse
Affiliation(s)
- Yili Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xiaodi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Limei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xiaoxiao Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Wenjia Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Qiong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| |
Collapse
|
30
|
Fernandes MGF, Pernin F, Antel JP, Kennedy TE. From BBB to PPP: Bioenergetic requirements and challenges for oligodendrocytes in health and disease. J Neurochem 2024. [PMID: 39253904 DOI: 10.1111/jnc.16219] [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: 03/31/2024] [Revised: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Mature myelinating oligodendrocytes, the cells that produce the myelin sheath that insulates axons in the central nervous system, have distinct energetic and metabolic requirements compared to neurons. Neurons require substantial energy to execute action potentials, while the energy needs of oligodendrocytes are directed toward building the lipid-rich components of myelin and supporting neuronal metabolism by transferring glycolytic products to axons as additional fuel. The utilization of energy metabolites in the brain parenchyma is tightly regulated to meet the needs of different cell types. Disruption of the supply of metabolites can lead to stress and oligodendrocyte injury, contributing to various neurological disorders, including some demyelinating diseases. Understanding the physiological properties, structures, and mechanisms involved in oligodendrocyte energy metabolism, as well as the relationship between oligodendrocytes and neighboring cells, is crucial to investigate the underlying pathophysiology caused by metabolic impairment in these disorders. In this review, we describe the particular physiological properties of oligodendrocyte energy metabolism and the response of oligodendrocytes to metabolic stress. We delineate the relationship between oligodendrocytes and other cells in the context of the neurovascular unit, and the regulation of metabolite supply according to energetic needs. We focus on the specific bioenergetic requirements of oligodendrocytes and address the disruption of metabolic energy in demyelinating diseases. We encourage further studies to increase understanding of the significance of metabolic stress on oligodendrocyte injury, to support the development of novel therapeutic approaches for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Milton Guilherme Forestieri Fernandes
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Florian Pernin
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jack P Antel
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Timothy E Kennedy
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
32
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
33
|
Netzahualcoyotzi C, Santillán-Cigales JJ, Adalid-Peralta LV, Velasco I. Infiltration of immune cells to the brain and its relation to the pathogenesis of Alzheimer's and Parkinson's diseases. J Neurochem 2024; 168:2316-2334. [PMID: 38549444 DOI: 10.1111/jnc.16106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 10/04/2024]
Abstract
The neurovascular unit, composed of vascular endothelium, vascular smooth muscle, extracellular matrix components, pericytes, astrocytes, microglia, and neurons, allows the highly regulated exchange of molecules and the limited trafficking of cells to the brain through coordinated signaling activity. The passage of peripheral immune cells to the brain parenchyma is observed when there is clear damage to the barriers of this neurovascular unit, as occurs in traumatic brain injury. The possibility of leukocyte infiltration to the brain in neurodegenerative conditions has been proposed. In this review, we focus on describing the evidence for peripheral immune cell infiltration to the brain in the two most frequent neurodegenerative diseases: Alzheimer's and Parkinson's diseases. In particular, we address the mechanisms that promote the passage of these cells into the brain under such pathological conditions. We also discuss the relevance of the resulting cellular interactions, which provide evidence that the presence of peripheral immune cells in the brain is a key point in these neurodegenerative diseases.
Collapse
Affiliation(s)
- Citlalli Netzahualcoyotzi
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Juan Jair Santillán-Cigales
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Virginia Adalid-Peralta
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| |
Collapse
|
34
|
Li H, Li Y, Zhong Q, Chen F, Wang H, Li X, Xie Y, Wang X. Dysfunction of neurovascular coupling in patients with cerebral small vessel disease: A combined resting-state fMRI and arterial spin labeling study. Exp Gerontol 2024; 194:112478. [PMID: 38866193 DOI: 10.1016/j.exger.2024.112478] [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: 04/15/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) closely correlates to cognitive impairment, but its pathophysiology and the neurovascular mechanisms of cognitive deficits were unclear. We aimed to explore the dysfunctional patterns of neurovascular coupling (NVC) in patients with CSVD and further investigate the neurovascular mechanisms of CSVD-related cognitive impairment. METHODS Forty-three patients with CSVD and twenty-four healthy controls were recruited. We adopted resting-state functional magnetic resonance imaging combined with arterial spin labeling to investigate the NVC dysfunctional patterns in patients with CSVD. The Human Brain Atlas with 246 brain regions was applied to extract the NVC coefficients for each brain region. Partial correlation analysis and mediation analysis were used to explore the relationship between CSVD pathological features, NVC dysfunctional patterns, and cognitive decline. RESULTS 8 brain regions with NVC dysfunction were found in patients with CSVD (p < 0.025, Bonferroni correction). The NVC dysfunctional patterns in regions of the default mode network and subcortical nuclei were negatively associated with lacunes, white matter hyperintensities burden, and the severity of CSVD (FDR correction, q < 0.05). The NVC decoupling in regions located in the default mode network positively correlated with delayed recall deficits (FDR correction, q < 0.05). Mediation analysis suggested that the decreased NVC pattern of the left superior frontal gyrus partially mediated the impact of white matter hyperintensities on delayed recall (Mediation effect: -0.119; 95%CI: -11.604,-0.458; p < 0.05). CONCLUSION The findings of this study reveal the NVC dysfunctional pattern in patients with CSVD and illustrate the neurovascular mechanism of CSVD-related cognitive impairment. The NVC function in the left superior frontal gyrus may serve as a promising biomarker and therapeutic target for memory deficits in patients with CSVD.
Collapse
Affiliation(s)
- Hui Li
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - You Li
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Qin Zhong
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Faxiang Chen
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Hui Wang
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Xiang Li
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Yuanliang Xie
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China.
| | - Xiang Wang
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China.
| |
Collapse
|
35
|
Rizzuti M, Melzi V, Brambilla L, Quetti L, Sali L, Ottoboni L, Meneri M, Ratti A, Verde F, Ticozzi N, Comi GP, Corti S, Abati E. Shaping the Neurovascular Unit Exploiting Human Brain Organoids. Mol Neurobiol 2024; 61:6642-6657. [PMID: 38334812 PMCID: PMC11338975 DOI: 10.1007/s12035-024-03998-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Brain organoids, three-dimensional cell structures derived from pluripotent stem cells, closely mimic key aspects of the human brain in vitro, providing a powerful tool for studying neurodevelopment and disease. The neuroectodermal induction protocol employed for brain organoid generation primarily gives rise to the neural cellular component but lacks the vital vascular system, which is crucial for the brain functions by regulating differentiation, migration, and circuit formation, as well as delivering oxygen and nutrients. Many neurological diseases are caused by dysfunctions of cerebral microcirculation, making vascularization of human brain organoids an important tool for pathogenetic and translational research. Experimentally, the creation of vascularized brain organoids has primarily focused on the fusion of vascular and brain organoids, on organoid transplantation in vivo, and on the use of microfluidic devices to replicate the intricate microenvironment of the human brain in vitro. This review summarizes these efforts and highlights the importance of studying the neurovascular unit in a forward-looking perspective of leveraging their use for understanding and treating neurological disorders.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linda Ottoboni
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Nicola Ticozzi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
36
|
Yu Z, Zhang H, Li L, Li Z, Chen D, Pang X, Ji Y, Wang Y. Microglia-mediated pericytes migration and fibroblast transition via S1P/S1P3/YAP signaling pathway after spinal cord injury. Exp Neurol 2024; 379:114864. [PMID: 38866101 DOI: 10.1016/j.expneurol.2024.114864] [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: 03/15/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
Platelet-derived growth factor receptor β positive (PDGFRβ+) pericytes detach from the microvascular wall and migrate into the injury center following spinal cord injury (SCI), which has been widely regarded as the main source of fibrotic scar, but the mechanism of migration and fibroblast transition remains elusive. Here we show the associated spatiotemporal distribution between microglia and pericytes at three and seven days post-injury (dpi). The increased expression of Sphingosine kinase-1 (SPHK1) in microglia significantly raised the concentration of Sphingosine-1-phosphate (S1P) in the spinal cord, which promotes migration and fibroblast transition of pericyte. In vitro experiments, we found the elevated Sphingosine 1-phosphate receptor 3 (S1P3), the S1P/S1PR3 axis inhibited the phosphorylation of YAP and promoted its nuclear translocation, which contributed to the formation of alpha-smooth muscle actin (α-SMA) and collagen type I (COL1) protein, This process can be blocked by an S1P3 specific inhibitor TY52156 in vitro. The S1P/S1P3/YAP pathway might be a potential target for treatment in SCI.
Collapse
Affiliation(s)
- Ziyuan Yu
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Huabin Zhang
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Linxi Li
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Zhi Li
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Danmin Chen
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Xiao Pang
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Yunxiang Ji
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Yezhong Wang
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China.
| |
Collapse
|
37
|
Chen A, Volpato G, Pong A, Schofield E, Huang J, Qiu Z, Paxinos G, Liang H. The Blood-Brain Barrier in Both Humans and Rats: A Perspective From 3D Imaging. Int J Biomed Imaging 2024; 2024:4482931. [PMID: 39224835 PMCID: PMC11368551 DOI: 10.1155/2024/4482931] [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: 07/19/2023] [Revised: 04/24/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Background: The blood-brain barrier (BBB) is part of the neurovascular unit (NVU) which plays a key role in maintaining homeostasis. However, its 3D structure is hardly known. The present study is aimed at imaging the BBB using tissue clearing and 3D imaging techniques in both human brain tissue and rat brain tissue. Methods: Both human and rat brain tissue were cleared using the CUBIC technique and imaged with either a confocal or two-photon microscope. Image stacks were reconstructed using Imaris. Results: Double staining with various antibodies targeting endothelial cells, basal membrane, pericytes of blood vessels, microglial cells, and the spatial relationship between astrocytes and blood vessels showed that endothelial cells do not evenly express CD31 and Glut1 transporter in the human brain. Astrocytes covered only a small portion of the vessels as shown by the overlap between GFAP-positive astrocytes and Collagen IV/CD31-positive endothelial cells as well as between GFAP-positive astrocytes and CD146-positive pericytes, leaving a big gap between their end feet. A similar structure was observed in the rat brain. Conclusions: The present study demonstrated the 3D structure of both the human and rat BBB, which is discrepant from the 2D one. Tissue clearing and 3D imaging are promising techniques to answer more questions about the real structure of biological specimens.
Collapse
Affiliation(s)
- Aiwen Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative MedicineShanghai Fourth People's HospitalSchool of MedicineTongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like IntelligenceShanghai Fourth People's HospitalSchool of MedicineTongji University, Shanghai, China
- Department of Anesthesiology and Perioperative MedicineShanghai Fourth People's HospitalSchool of MedicineTongji University, Shanghai, China
- Department of AcupunctureShuguang HospitalShanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gavin Volpato
- Department of Brain Structure and FunctionNeuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical SciencesThe University of New South Wales, Kensington, New South Wales, Australia
| | - Alice Pong
- Department of Brain Structure and FunctionNeuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical SciencesThe University of New South Wales, Kensington, New South Wales, Australia
| | - Emma Schofield
- Department of Brain Structure and FunctionNeuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical SciencesThe University of New South Wales, Kensington, New South Wales, Australia
| | - Jun Huang
- School of Chemical and Biomolecular EngineeringThe University of Sydney, Camperdown, New South Wales, Australia
| | - Zizhao Qiu
- Centre of Life ScienceSuzhou Industrial Park Monash Research Institute of Science and TechnologySoutheast University-Monash University Joint Graduate SchoolMonash University-Southeast University Joint Research Institute, Suzhou, Jiangsu Province, China
| | - George Paxinos
- Department of Brain Structure and FunctionNeuroscience Research Australia, Randwick, New South Wales, Australia
- School of Medical SciencesThe University of New South Wales, Kensington, New South Wales, Australia
| | - Huazheng Liang
- Translational Research Institute of Brain and Brain-Like IntelligenceShanghai Fourth People's HospitalSchool of MedicineTongji University, Shanghai, China
- Department of Brain Structure and FunctionNeuroscience Research Australia, Randwick, New South Wales, Australia
- Centre of Life ScienceSuzhou Industrial Park Monash Research Institute of Science and TechnologySoutheast University-Monash University Joint Graduate SchoolMonash University-Southeast University Joint Research Institute, Suzhou, Jiangsu Province, China
| |
Collapse
|
38
|
Hainsworth AH, Blackburn TP, Bradshaw EM, Elahi FM, Gorelick PB, Isaacs JD, Wallin A, Williams SC. The promise of molecular science in brain health. What breakthroughs are anticipated in the next 20 years? CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 7:100364. [PMID: 39263555 PMCID: PMC11387710 DOI: 10.1016/j.cccb.2024.100364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/28/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Brain health means optimal physiological brain function across the normal life-course. It encompasses not only healthy brain aging but also brain diseases, their diagnosis and treatment. In all these areas, molecular science has advanced our understanding. This multi-disciplinary review combines viewpoints from laboratory science, clinical medicine and the bioscience industry. First, we review the advances that molecular science has brought to brain health in the past twenty years. These include therapeutic antibodies for CNS diseases (multiple sclerosis, Alzheimer disease) and the dramatic introduction of RNA-targeted therapeutics. Second, we highlight areas where greater molecular understanding is needed. Salient examples are the relation of brain structure to cognitive symptoms, and molecular biomarkers for diagnosis, target discovery and testing of interventions. Finally, we speculate on aspects of molecular science that are likely to advance brain health in the next twenty years. These include: cell senescence and chronobiology; gene editing (notably, CRISPR) and RNA targeting (RNA interference, miRNA manipulation); brain-immune interactions; novel drug targets (AQP4, HIF1, Toll-like receptors); and novel chemistry to make new drugs (molecular machines, quantum molecular modelling and "click" chemistry). Early testing of the relationships between molecular pathways and clinical manifestations will drive much-needed breakthroughs in neurology and psychiatry.
Collapse
Affiliation(s)
- Atticus H Hainsworth
- Molecular & Clinical Sciences Research Institute, St George's University of London, London, SW17 0RE, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, SW17 0QT, UK
| | - Thomas P Blackburn
- Translational Pharmacology BioVentures, Leigh on Sea, Essex, SS9 2UA, UK
- TPBioVentures, Hoboken, NJ, USA
| | - Elizabeth M Bradshaw
- Carol and Gene Ludwig Center for Research on Neurodegeneration, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Fanny M Elahi
- Departments of Neurology and Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029-5674, USA
- James J. Peter VA Medical Center, Bronx, NY, USA
| | - Philip B Gorelick
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine, 635 N. Michigan Avenue, Chicago, IL 60611, USA
| | - Jeremy D Isaacs
- Molecular & Clinical Sciences Research Institute, St George's University of London, London, SW17 0RE, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, SW17 0QT, UK
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Steven Cr Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, Kings College London. SE5 8AF, UK
| |
Collapse
|
39
|
Villareal JAB, Bathe T, Hery GP, Phillips JL, Tsering W, Prokop S. Deterioration of neuroimmune homeostasis in Alzheimer's Disease patients who survive a COVID-19 infection. J Neuroinflammation 2024; 21:202. [PMID: 39154174 PMCID: PMC11330027 DOI: 10.1186/s12974-024-03196-3] [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: 05/21/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Growing evidence has implicated systemic infection as a significant risk factor for the development and advancement of Alzheimer's disease (AD). With the emergence of SARS-CoV-2 (COVID-19) and the resultant pandemic, many individuals from the same aging population vulnerable to AD suffered a severe systemic infection with potentially unidentified long-term consequences for survivors. To study the impact of COVID-19 survival on the brain's intrinsic immune system in a population also suffering from AD, we profiled post-mortem brain tissue from patients in the UF Neuromedicine Human Brain and Tissue Bank with a diagnosis of AD who survived a COVID-19 infection (COVID-AD) and contrasted our findings with AD patients who did not experience a COVID-19 infection, including a group of brain donors who passed away before arrival of SARS-CoV-2 in the United States. We assessed disease-relevant protein pathology and microglial and astrocytic markers by quantitative immunohistochemistry and supplemented these data with whole tissue gene expression analysis performed on the NanoString nCounter® platform. COVID-AD patients showed slightly elevated Aβ burden in the entorhinal, fusiform, and inferior temporal cortices compared to non-COVID-AD patients, while tau pathology burden did not differ between groups. Analysis of microglia revealed a significant loss of microglial homeostasis as well as exacerbated microgliosis in COVID-AD patients compared to non-COVID-AD patients in a brain region-dependent manner. Furthermore, COVID-AD patients showed reduced cortical astrocyte numbers, independent of functional subtype. Transcriptomic analysis supported these histological findings and, in addition, identified a dysregulation of oligodendrocyte and myelination pathways in the hippocampus of COVID-AD patients. In summary, our data demonstrate a profound impact of COVID-19 infection on neuroimmune and glial pathways in AD patients persisting for months post-infection, highlighting the importance of peripheral to central neuroimmune crosstalk in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan A B Villareal
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Tim Bathe
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Gabriela P Hery
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Jennifer L Phillips
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Wangchen Tsering
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32608, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
40
|
Delgado-García LM, Ojalvo-Sanz AC, Nakamura TKE, Martín-López E, Porcionatto M, Lopez-Mascaraque L. Dissecting reactive astrocyte responses: lineage tracing and morphology-based clustering. Biol Res 2024; 57:54. [PMID: 39143594 PMCID: PMC11323641 DOI: 10.1186/s40659-024-00532-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Brain damage triggers diverse cellular and molecular events, with astrocytes playing a crucial role in activating local neuroprotective and reparative signaling within damaged neuronal circuits. Here, we investigated reactive astrocytes using a multidimensional approach to categorize their responses into different subtypes based on morphology. This approach utilized the StarTrack lineage tracer, single-cell imaging reconstruction and multivariate data analysis. Our findings identified three profiles of reactive astrocyte responses, categorized by their effects on cell size- and shape- related morphological parameters: "moderate", "strong," and "very strong". We also examined the heterogeneity of astrocyte reactivity, focusing on spatial and clonal distribution. Our research revealed a notable enrichment of protoplasmic and fibrous astrocytes within the "strong" and "very strong" response subtypes. Overall, our study contributes to a better understanding of astrocyte heterogeneity in response to an injury. By characterizing the diverse reactive responses among astrocyte subpopulations, we provide insights that could guide future research aimed at identifying novel therapeutic targets to mitigate brain damage and promote neural repair.
Collapse
Affiliation(s)
- Lina M Delgado-García
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal-CSIC, Avenida Dr. Arce 37, 28002, Madrid, Spain
- Departamento de Bioquímica, Universidade Federal de Sao Paulo UNIFESP, Rua Pedro de Toledo 669, Sao Paulo, 04039032, Brazil
| | - Ana C Ojalvo-Sanz
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal-CSIC, Avenida Dr. Arce 37, 28002, Madrid, Spain
| | - Thabatta K E Nakamura
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal-CSIC, Avenida Dr. Arce 37, 28002, Madrid, Spain
- Departamento de Bioquímica, Universidade Federal de Sao Paulo UNIFESP, Rua Pedro de Toledo 669, Sao Paulo, 04039032, Brazil
| | - Eduardo Martín-López
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal-CSIC, Avenida Dr. Arce 37, 28002, Madrid, Spain
- Departments of Neurosurgery and Neuroscience, Yale University School of Medicine, 310 Cedar Street, New Haven, CT, 06510, USA
| | - Marimelia Porcionatto
- Departamento de Bioquímica, Universidade Federal de Sao Paulo UNIFESP, Rua Pedro de Toledo 669, Sao Paulo, 04039032, Brazil
| | - Laura Lopez-Mascaraque
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal-CSIC, Avenida Dr. Arce 37, 28002, Madrid, Spain.
| |
Collapse
|
41
|
Ewees MG, El-Mahdy MA, Hannawi Y, Zweier JL. Tobacco cigarette smoking induces cerebrovascular dysfunction followed by oxidative neuronal injury with the onset of cognitive impairment. J Cereb Blood Flow Metab 2024:271678X241270415. [PMID: 39136181 DOI: 10.1177/0271678x241270415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
While chronic smoking triggers cardiovascular disease, controversy remains regarding its effects on the brain and cognition. We investigated the effects of long-term cigarette smoke (CS) exposure (CSE) on cerebrovascular function, neuronal injury, and cognition in a novel mouse exposure model. Longitudinal studies were performed in CS or air-exposed mice, 2 hours/day, for up to 60 weeks. Hypertension and carotid vascular endothelial dysfunction (VED) occurred by 16 weeks of CSE, followed by reduced carotid artery blood flow, with oxidative stress detected in the carotid artery, and subsequently in the brain of CS-exposed mice with generation of reactive oxygen species (ROS) and secondary protein and DNA oxidation, microglial activation and astrocytosis. Brain small vessels exhibited decreased levels of endothelial NO synthase (eNOS), enlarged perivascular spaces with blood brain barrier (BBB) leak and decreased levels of tight-junction proteins. In the brain, amyloid-β deposition and phosphorylated-tau were detected with increases out to 60 weeks, at which time mice exhibited impaired spatial learning and memory. Thus, long-term CSE initiates a cascade of ROS generation and oxidative damage, eNOS dysfunction with cerebral hypoperfusion, as well as cerebrovascular and BBB damage with intracerebral inflammation, and neuronal degeneration, followed by the onset of impaired cognition and memory.
Collapse
Affiliation(s)
- Mohamed G Ewees
- Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Mohamed A El-Mahdy
- Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jay L Zweier
- Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
42
|
Chen H, Mirg S, Gaddale P, Agrawal S, Li M, Nguyen V, Xu T, Li Q, Liu J, Tu W, Liu X, Drew PJ, Zhang N, Gluckman BJ, Kothapalli S. Multiparametric Brain Hemodynamics Imaging Using a Combined Ultrafast Ultrasound and Photoacoustic System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401467. [PMID: 38884161 PMCID: PMC11336909 DOI: 10.1002/advs.202401467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/25/2024] [Indexed: 06/18/2024]
Abstract
Studying brain-wide hemodynamic responses to different stimuli at high spatiotemporal resolutions can help gain new insights into the mechanisms of neuro- diseases and -disorders. Nonetheless, this task is challenging, primarily due to the complexity of neurovascular coupling, which encompasses interdependent hemodynamic parameters including cerebral blood volume (CBV), cerebral blood flow (CBF), and cerebral oxygen saturation (SO2). The current brain imaging technologies exhibit inherent limitations in resolution, sensitivity, and imaging depth, restricting their capacity to comprehensively capture the intricacies of cerebral functions. To address this, a multimodal functional ultrasound and photoacoustic (fUSPA) imaging platform is reported, which integrates ultrafast ultrasound and multispectral photoacoustic imaging methods in a compact head-mountable device, to quantitatively map individual dynamics of CBV, CBF, and SO2 as well as contrast agent enhanced brain imaging at high spatiotemporal resolutions. Following systematic characterization, the fUSPA system is applied to study brain-wide cerebrovascular reactivity (CVR) at single-vessel resolution via relative changes in CBV, CBF, and SO2 in response to hypercapnia stimulation. These results show that cortical veins and arteries exhibit differences in CVR in the stimulated state and consistent anti-correlation in CBV oscillations during the resting state, demonstrating the multiparametric fUSPA system's unique capabilities in investigating complex mechanisms of brain functions.
Collapse
Affiliation(s)
- Haoyang Chen
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Shubham Mirg
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Prameth Gaddale
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Sumit Agrawal
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Menghan Li
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Van Nguyen
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Tianbao Xu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Qiong Li
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Jinyun Liu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Wenyu Tu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Xiao Liu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Institute for Computational and Data SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Patrick J. Drew
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Engineering Science and MechanicsThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of BiologyThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of NeurosurgeryThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Nanyin Zhang
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Bruce J. Gluckman
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Engineering Science and MechanicsThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of NeurosurgeryThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Sri‐Rajasekhar Kothapalli
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Penn State Cancer InstituteThe Pennsylvania State UniversityHersheyPA17033USA
- Graduate Program in AcousticsThe Pennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
43
|
Ryman SG, Vakhtin AA, Mayer AR, van der Horn HJ, Shaff NA, Nitschke SR, Julio KR, Tarawneh RM, Rosenberg GA, Diaz SV, Pirio Richardson SE, Lin HC. Abnormal Cerebrovascular Activity, Perfusion, and Glymphatic Clearance in Lewy Body Diseases. Mov Disord 2024; 39:1258-1268. [PMID: 38817039 PMCID: PMC11341260 DOI: 10.1002/mds.29867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024] Open
Abstract
Cerebrovascular activity is not only crucial to optimal cerebral perfusion, but also plays an important role in the glymphatic clearance of interstitial waste, including α-synuclein. This highlights a need to evaluate how cerebrovascular activity is altered in Lewy body diseases. This review begins by discussing how vascular risk factors and cardiovascular autonomic dysfunction may serve as upstream or direct influences on cerebrovascular activity. We then discuss how patients with Lewy body disease exhibit reduced and delayed cerebrovascular activity, hypoperfusion, and reductions in measures used to capture cerebrospinal fluid flow, suggestive of a reduced capacity for glymphatic clearance. Given the lack of an existing framework, we propose a model by which these processes may foster α-synuclein aggregation and neuroinflammation. Importantly, this review highlights several avenues for future research that may lead to treatments early in the disease course, prior to neurodegeneration. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Andrei A Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Andrew R Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Harm Jan van der Horn
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Nicholas A Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Stephanie R Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Kayla R Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Rawan M Tarawneh
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
- Cognitive Neurology Section, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Gary A Rosenberg
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Shanna V Diaz
- Department of Internal Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Sarah E Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
- New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| | - Henry C Lin
- Department of Internal Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
- New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| |
Collapse
|
44
|
Lang AN, Zhong Y, Lei W, Xiao Y, Hang Y, Xie Y, Lv Z, Zhang Y, Liu X, Liang M, Zhang C, Zhang P, Yang H, Wu Y, Wang Q, Yang K, Long J, Liu Y, Wang S, Tang Y, Lei M, Zhang D, Ouyang L, Zhang L, Wang C. Neural mechanism of non-adaptive cognitive emotion regulation in patients with non-suicidal self-injury. Compr Psychiatry 2024; 133:152487. [PMID: 38714144 DOI: 10.1016/j.comppsych.2024.152487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/22/2024] [Accepted: 04/13/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The incidence of non-suicidal self-injury (NSSI) has been on the rise in recent years. Studies have shown that people with NSSI have difficulties in emotion regulation and cognitive control. In addition, some studies have investigated the cognitive emotion regulation of people with NSSI which found that they have difficulties in cognitive emotion regulation, but there was a lack of research on cognitive emotion regulation strategies and related neural mechanisms. METHODS This study included 117 people with NSSI (age = 19.47 ± 5.13, male = 17) and 84 non-NSSI participants (age = 19.86 ± 4.14, male = 16). People with NSSI met the DSM-5 diagnostic criteria, and non-NSSI participants had no mental or physical disorders. The study collected all participants' data of Cognitive Emotion Regulation Questionnaire (CERQ) and functional magnetic resonance imaging (fMRI) to explore the differences in psychological performance and brain between two groups. Afterwards, Machine learning was used to select the found differential brain regions to obtain the highest correlation regions with NSSI. Then, Allen's Human Brain Atlas database was used to compare with the information on the abnormal brain regions of people with NSSI to find the genetic information related to NSSI. In addition, gene enrichment analysis was carried out to find the related pathways and specific cells that may have differences. RESULTS The differences between NSSI participants and non-NSSI participants were as follows: positive refocusing (t = -4.74, p < 0.01); refocusing on plans (t = -4.11, p < 0.01); positive reappraisal (t = -9.22, p < 0.01); self-blame (t = 6.30, p < 0.01); rumination (t = 3.64, p < 0.01); catastrophizing (t = 9.10, p < 0.01), and blaming others (t = 2.52, p < 0.01), the precentral gyrus (t = 6.04, pFDR < 0.05) and the rolandic operculum (t = -4.57, pFDR < 0.05). Rolandic operculum activity was negatively correlated with blaming others (r = -0.20, p < 0.05). Epigenetic results showed that excitatory neurons (p < 0.01) and inhibitory neurons (p < 0.01) were significant differences in two pathways, "trans-synaptic signaling" (p < -log108) and "modulation of chemical synaptic transmission" (p < -log108) in both cells. CONCLUSIONS People with NSSI are more inclined to adopt non-adaptive cognitive emotion regulation strategies. Rolandic operculum is also abnormally active. Abnormal changes in the rolandic operculum of them are associated with non-adaptive cognitive emotion regulation strategies. Changes in the excitatory and inhibitory neurons provide hints to explore the abnormalities of the neurological mechanisms at the cellular level of them. Trial registration number NCT04094623.
Collapse
Affiliation(s)
- Author Nan Lang
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China; The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Yuan Zhong
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Mental Health and Cognitive Science, Nanjing Normal University, Nanjing, China
| | - Wenkun Lei
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Yiwen Xiao
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Yaming Hang
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China; The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Ya Xie
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Zhangwei Lv
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Yumin Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Xinyao Liu
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Minlu Liang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Congjie Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Pei Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Hua Yang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Yun Wu
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Qiuyu Wang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Kun Yang
- The Third Hospital of Mianyang, 190 Jiannan Road, Youxian District, Mianyang, Sichuan, China
| | - Jing Long
- Tianjin Anding Hospital, 13 Liulin Road, Hexi District, Tianjin, China
| | - Yuan Liu
- Xuzhou Oriental People's Hospital, 379 Dongdianzitongshan Road, Yunlong District, Xuzhou, Jiangsu, China
| | - Suhong Wang
- The First People's Hospital of Changzhou, 185 Juqian Road, Tianning District, Changzhou, Jiangsu, China
| | - Yibin Tang
- College of Internet of Things Engineering, Hohai University, Changzhou, Jiangsu, China
| | - Maochun Lei
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China; The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Danyu Zhang
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China; The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Lichen Ouyang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Liping Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China
| | - Chun Wang
- School of Psychology, Nanjing Normal University, Nanjing, Jiangsu, China; The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Gulou District, Nanjing, Jiangsu, China; Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
45
|
Rathbone E, Fu D. Quantitative Optical Imaging of Oxygen in Brain Vasculature. J Phys Chem B 2024; 128:6975-6989. [PMID: 38991095 DOI: 10.1021/acs.jpcb.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
The intimate relationship between neuronal activity and cerebral oxygenation underpins fundamental brain functions like cognition, sensation, and motor control. Optical imaging offers a noninvasive approach to assess brain oxygenation and often serves as an indirect proxy for neuronal activity. However, deciphering neurovascular coupling─the intricate interplay between neuronal activity, blood flow, and oxygen delivery─necessitates independent, high spatial resolution, and high temporal resolution measurements of both microvasculature oxygenation and neuronal activation. This Perspective examines the established optical techniques employed for brain oxygen imaging, specifically functional near-infrared spectroscopy, photoacoustic imaging, optical coherence tomography, and two-photon phosphorescent lifetime microscopy, highlighting their fundamental principles, strengths, and limitations. Several other emerging optical techniques are also introduced. Finally, we discuss key technological challenges and future directions for quantitative optical oxygen imaging, paving the way for a deeper understanding of oxygen metabolism in the brain.
Collapse
Affiliation(s)
- Emily Rathbone
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
46
|
Chandra S, Vassar R. The role of the gut microbiome in the regulation of astrocytes in Alzheimer's disease. Neurotherapeutics 2024:e00425. [PMID: 39054180 DOI: 10.1016/j.neurot.2024.e00425] [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: 04/01/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and is the most common cause of dementia. AD is characterized pathologically by proteinaceous aggregates composed of amyloid beta (Aβ) and tau as well as progressive neurodegeneration. Concurrently with the buildup of protein aggregates, a strong neuroinflammatory response, in the form of reactive astrocytosis and microgliosis, occurs in the AD brain. It has recently been shown that the gut microbiome (GMB), composed of trillions of bacteria in the human intestine, can regulate both reactive astrocytosis and microgliosis in the context of both amyloidosis and tauopathy. Many studies have implicated microglia in these processes. However, growing evidence suggests that interactions between the GMB and astrocytes have a much larger role than previously thought. In this review, we summarize evidence regarding the gut microbiome in the control of reactive astrocytosis in AD.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA; Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Robert Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
47
|
Ye W, Xu S, Liu Y, Ye Z. Role of endothelial glycocalyx in central nervous system diseases and evaluation of the targeted therapeutic strategies for its protection: a review of clinical and experimental data. Rev Neurosci 2024; 0:revneuro-2024-0039. [PMID: 39034663 DOI: 10.1515/revneuro-2024-0039] [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: 03/14/2024] [Accepted: 05/22/2024] [Indexed: 07/23/2024]
Abstract
Central nervous system (CNS) diseases, such as stroke, traumatic brain injury, dementia, and demyelinating diseases, are generally characterized by high morbidity and mortality, which impose a heavy economic burden on patients and their caregivers throughout their lives as well as on public health. The occurrence and development of CNS diseases are closely associated with a series of pathophysiological changes including inflammation, blood-brain barrier disruption, and abnormal coagulation. Endothelial glycocalyx (EG) plays a key role in these changes, making it a novel intervention target for CNS diseases. Herein, we review the current understanding of the role of EG in common CNS diseases, from the perspective of individual pathways/cytokines in pathophysiological and systematic processes. Furthermore, we emphasize the recent developments in therapeutic agents targeted toward protection or restoration of EG. Some of these treatments have yielded unexpected pharmacological results, as previously unknown mechanisms underlying the degradation and destruction of EG has been brought to light. Furthermore, the anti-inflammatory, anticoagulative, and antioxidation effects of EG and its protective role exerted via the blood-brain barrier have been recognized.
Collapse
Affiliation(s)
- Weihao Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shang Xu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying Liu
- Department of Rehabilitation Medicine, 117742The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ziming Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
48
|
Nuszkiewicz J, Kukulska-Pawluczuk B, Piec K, Jarek DJ, Motolko K, Szewczyk-Golec K, Woźniak A. Intersecting Pathways: The Role of Metabolic Dysregulation, Gastrointestinal Microbiome, and Inflammation in Acute Ischemic Stroke Pathogenesis and Outcomes. J Clin Med 2024; 13:4258. [PMID: 39064298 PMCID: PMC11278353 DOI: 10.3390/jcm13144258] [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: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of mortality and long-term disability worldwide, driven by complex and multifaceted etiological factors. Metabolic dysregulation, gastrointestinal microbiome alterations, and systemic inflammation are emerging as significant contributors to AIS pathogenesis. This review addresses the critical need to understand how these factors interact to influence AIS risk and outcomes. We aim to elucidate the roles of dysregulated adipokines in obesity, the impact of gut microbiota disruptions, and the neuroinflammatory cascade initiated by lipopolysaccharides (LPS) in AIS. Dysregulated adipokines in obesity exacerbate inflammatory responses, increasing AIS risk and severity. Disruptions in the gut microbiota and subsequent LPS-induced neuroinflammation further link systemic inflammation to AIS. Advances in neuroimaging and biomarker development have improved diagnostic precision. Here, we highlight the need for a multifaceted approach to AIS management, integrating metabolic, microbiota, and inflammatory insights. Potential therapeutic strategies targeting these pathways could significantly improve AIS prevention and treatment. Future research should focus on further elucidating these pathways and developing targeted interventions to mitigate the impacts of metabolic dysregulation, microbiome imbalances, and inflammation on AIS.
Collapse
Affiliation(s)
- Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Beata Kukulska-Pawluczuk
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Katarzyna Piec
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Dorian Julian Jarek
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Karina Motolko
- Student Research Club of Neurology, Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland;
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| |
Collapse
|
49
|
Lambert T, Niknejad HR, Kil D, Brunner C, Nuttin B, Montaldo G, Urban A. Functional ultrasound imaging and neuronal activity: how accurate is the spatiotemporal match? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602912. [PMID: 39026833 PMCID: PMC11257620 DOI: 10.1101/2024.07.10.602912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Over the last decade, functional ultrasound (fUS) has risen as a critical tool in functional neuroimaging, leveraging hemodynamic changes to infer neural activity indirectly. Recent studies have established a strong correlation between neural spike rates (SR) and functional ultrasound signals. However, understanding their spatial distribution and variability across different brain areas is required to thoroughly interpret fUS signals. In this regard, we conducted simultaneous fUS imaging and Neuropixels recordings during stimulus-evoked activity in awake mice within three regions the visual pathway. Our findings indicate that the temporal dynamics of fUS and SR signals are linearly correlated, though the correlation coefficients vary among visual regions. Conversely, the spatial correlation between the two signals remains consistent across all regions with a spread of approximately 300 micrometers. Finally, we introduce a model that integrates the spatial and temporal components of the fUS signal, allowing for a more accurate interpretation of fUS images.
Collapse
Affiliation(s)
- Théo Lambert
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Hamid Reza Niknejad
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
- University Medical Center Utrecht, Utrecht, The Netherlands (current affiliation)
| | - Dries Kil
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Clément Brunner
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Bart Nuttin
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Gabriel Montaldo
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Alan Urban
- Neuro-Electronics Research Flanders, Leuven, Belgium
- VIB, Leuven, Belgium
- Imec, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Rigollet S, Rome C, Ador T, Dumont E, Pichon C, Delalande A, Barbier EL, Stupar V. FUS-mediated BBB opening leads to transient perfusion decrease and inflammation without acute or chronic brain lesion. Theranostics 2024; 14:4147-4160. [PMID: 38994025 PMCID: PMC11234282 DOI: 10.7150/thno.96721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Impact: The permeabilization of the BBB to deliver therapeutics with MR-guided FUS redefines therapeutic strategies as it improves patient outcomes. To ensure the best translation towards clinical treatment, the evaluation of hemodynamic modifications in the CNS is necessary to refine treatment parameters. Methods: MR-guided FUS was applied at 1.5 MHz with a 50 ms burst every 1 s to open the BBB. CBF, BVf and ADC parameters were monitored with MRI. Cavitation was monitored with a PCD during the FUS sequence and classified with the IUD index into three cavitation levels. We distinctly applied the FUS in the cortex or the striatum. After the BBB permeabilization, neuroinflammation markers were quantified longitudinally. Results: The BBB was successfully opened in all animals in this study and only one animal was classified as "hard" and excluded from the rest of the study. 30 min after FUS-induced BBB opening in the cortex, we measured a 54% drop in CBF and a 13% drop in BVf compared to the contralateral side. After permeabilization of the striatum, a 38% drop in CBF and a 15% drop in BVf were measured. CBF values rapidly returned to baseline, and 90 min after BBB opening, no significant differences were observed. We quantified the subsequent neuroinflammation, noting a significant increase in astrocytic recruitment at 2 days and microglial activation at 1 day after FUS. After 7 days, no more inflammation was visible in the brain. Conclusion: FUS-induced BBB opening transiently modifies hemodynamic parameters such as CBF and BVf, suggesting limited nutrients and oxygen supply to the CNS in the hour following the procedure.
Collapse
Affiliation(s)
- Sébastien Rigollet
- Image Guided Therapy, Pessac, France
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Thomas Ador
- Université d'Orléans, LI²RSO, Orléans, France
- ART ARNm, Inserm US55, Orléans, France
- Laboratory of Experimental and Molecular Immunology and Neuromodulation (INEM), UMR 7355 CNRS-University of Orleans, Orleans, France
| | | | - Chantal Pichon
- Université d'Orléans, LI²RSO, Orléans, France
- ART ARNm, Inserm US55, Orléans, France
- Institut Universitaire de France, Paris, France
| | - Anthony Delalande
- Université d'Orléans, LI²RSO, Orléans, France
- ART ARNm, Inserm US55, Orléans, France
| | - Emmanuel L. Barbier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CNRS, IRMaGe, Grenoble, France
| | - Vasile Stupar
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, CNRS, IRMaGe, Grenoble, France
| |
Collapse
|