51
|
Loo JH, Wang Z, Chong RS. Caveolin-1 in vascular health and glaucoma: A critical vascular regulator and potential therapeutic target. Front Med (Lausanne) 2023; 10:1087123. [PMID: 36760400 PMCID: PMC9902660 DOI: 10.3389/fmed.2023.1087123] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/04/2023] [Indexed: 01/25/2023] Open
Abstract
Caveolin-1 (Cav-1) is an integral scaffolding membrane protein found in most cell types. Cav-1 has been found to contribute significantly to ocular function, with mutations of Cav-1 being associated with a genetic risk of glaucoma development. Raised intraocular pressure (IOP) is a major modifiable risk factor for glaucoma. Cav-1 may be involved in both IOP-dependent and independent mechanisms involving vascular dysregulation. Systemic vascular diseases including hypertension, diabetes and hyperlipidaemia, have been shown to be associated with glaucoma development. Cav-1 is closely interlinked with endothelial nitric oxide synthase pathways that mediate vascular function and prevent cardiovascular diseases. Endothelial nitric oxide synthase and endothelin-1 are key vasoactive molecules expressed in retinal blood vessels that function to autoregulate ocular blood flow (OBF). Disruptions in the homeostasis of OBF have led to a growing concept of impaired neurovascular coupling in glaucoma. The imbalance between perfusion and neuronal stimulation arising from Cav-1 depletion may result in relative ischemia of the optic nerve head and glaucomatous injury. OBF is also governed by circadian variation in IOP and systemic blood pressure (BP). Cav-1 has been shown to influence central BP variability and other circadian rhythms such as the diurnal phagolysosomal digestion of photoreceptor fragments and toxic substrates to maintain ocular health. Overall, the vast implications of Cav-1 on various ocular mechanisms leading to glaucoma suggest a potential for new therapeutics to enhance Cav-1 expression, which has seen success in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Hong Loo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Rachel S. Chong
- Glaucoma Department, Singapore National Eye Center, Singapore, Singapore,Ocular Imaging Department, Singapore Eye Research Institute, Singapore, Singapore,*Correspondence: Rachel S. Chong ✉
| |
Collapse
|
52
|
Reiss Y, Bauer S, David B, Devraj K, Fidan E, Hattingen E, Liebner S, Melzer N, Meuth SG, Rosenow F, Rüber T, Willems LM, Plate KH. The neurovasculature as a target in temporal lobe epilepsy. Brain Pathol 2023; 33:e13147. [PMID: 36599709 PMCID: PMC10041171 DOI: 10.1111/bpa.13147] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The blood-brain barrier (BBB) is a physiological barrier maintaining a specialized brain micromilieu that is necessary for proper neuronal function. Endothelial tight junctions and specific transcellular/efflux transport systems provide a protective barrier against toxins, pathogens, and immune cells. The barrier function is critically supported by other cell types of the neurovascular unit, including pericytes, astrocytes, microglia, and interneurons. The dysfunctionality of the BBB is a hallmark of neurological diseases, such as ischemia, brain tumors, neurodegenerative diseases, infections, and autoimmune neuroinflammatory disorders. Moreover, BBB dysfunction is critically involved in epilepsy, a brain disorder characterized by spontaneously occurring seizures because of abnormally synchronized neuronal activity. While resistance to antiseizure drugs that aim to reduce neuronal hyperexcitability remains a clinical challenge, drugs targeting the neurovasculature in epilepsy patients have not been explored. The use of novel imaging techniques permits early detection of BBB leakage in epilepsy; however, the detailed mechanistic understanding of causes and consequences of BBB compromise remains unknown. Here, we discuss the current knowledge of BBB involvement in temporal lobe epilepsy with the emphasis on the neurovasculature as a therapeutic target.
Collapse
Affiliation(s)
- Yvonne Reiss
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Sebastian Bauer
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Bastian David
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Kavi Devraj
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Elif Fidan
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Elke Hattingen
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Institute of Neuroradiology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| | - Nico Melzer
- Department of Neurology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Felix Rosenow
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Theodor Rüber
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany.,Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Laurent M Willems
- Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany.,Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Center of Neurology and Neurosurgery, University Hospital, Goethe University, Frankfurt, Germany
| | - Karl H Plate
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), University Hospital, Goethe University, Frankfurt, Germany
| |
Collapse
|
53
|
Preishuber-Pflügl J, Mayr D, Altinger V, Brunner SM, Koller A, Runge C, Ladek AM, Lenzhofer M, Rivera FJ, Tempfer H, Aigner L, Reitsamer HA, Trost A. Pericyte-derived cells participate in optic nerve scar formation. Front Physiol 2023; 14:1151495. [PMID: 37143930 PMCID: PMC10151493 DOI: 10.3389/fphys.2023.1151495] [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: 01/26/2023] [Accepted: 04/04/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction: Pericytes (PCs) are specialized cells located abluminal of endothelial cells on capillaries, fulfilling numerous important functions. Their potential involvement in wound healing and scar formation is achieving increasing attention since years. Thus, many studies investigated the participation of PCs following brain and spinal cord (SC) injury, however, lacking in-depth analysis of lesioned optic nerve (ON) tissue. Further, due to the lack of a unique PC marker and uniform definition of PCs, contradicting results are published. Methods: In the present study the inducible PDGFRβ-P2A-CreERT2-tdTomato lineage tracing reporter mouse was used to investigate the participation and trans-differentiation of endogenous PC-derived cells in an ON crush (ONC) injury model, analyzing five different post lesion time points up to 8 weeks post lesion. Results: PC-specific labeling of the reporter was evaluated and confirmed in the unlesioned ON of the reporter mouse. After ONC, we detected PC-derived tdTomato+ cells in the lesion, whereof the majority is not associated with vascular structures. The number of PC-derived tdTomato+ cells within the lesion increased over time, accounting for 60-90% of all PDGFRβ+ cells in the lesion. The presence of PDGFRβ+tdTomato- cells in the ON scar suggests the existence of fibrotic cell subpopulations of different origins. Discussion: Our results clearly demonstrate the presence of non-vascular associated tdTomato+ cells in the lesion core, indicating the participation of PC-derived cells in fibrotic scar formation following ONC. Thus, these PC-derived cells represent promising target cells for therapeutic treatment strategies to modulate fibrotic scar formation to improve axonal regeneration.
Collapse
Affiliation(s)
- Julia Preishuber-Pflügl
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Daniela Mayr
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Veronika Altinger
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Susanne M. Brunner
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Andreas Koller
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Christian Runge
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Anja-Maria Ladek
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Markus Lenzhofer
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Francisco J. Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Translational Regenerative Neurobiology Group, Molecular and Integrative Biosciences Research Program (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Herbert Tempfer
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Herbert A. Reitsamer
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Austria
- Director of the Research Program for Experimental Ophthalmology and Glaucoma Research, Salzburg, Austria
| | - Andrea Trost
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- *Correspondence: Andrea Trost,
| |
Collapse
|
54
|
Davis CM, Ibrahim AH, Alkayed NJ. Cytochrome P450-derived eicosanoids in brain: From basic discovery to clinical translation. ADVANCES IN PHARMACOLOGY 2023; 97:283-326. [DOI: 10.1016/bs.apha.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
55
|
Hattori Y. The Multiple Roles of Pericytes in Vascular Formation and Microglial Functions in the Brain. Life (Basel) 2022; 12:1835. [PMID: 36362989 PMCID: PMC9699346 DOI: 10.3390/life12111835] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 10/15/2023] Open
Abstract
In the capillary walls, vascular endothelial cells are covered with mural cells, such as smooth muscle cells and pericytes. Although pericytes had been thought to play simply a structural role, emerging evidence has highlighted their multiple functions in the embryonic, postnatal, and adult brain. As the central nervous system (CNS) develops, the brain's vascular structure gradually matures into a hierarchical network, which is crucial for the proper development of neural lineage cells by providing oxygen and nutrients. Pericytes play an essential role in vascular formation and regulate blood‒brain barrier (BBB) integrity as a component of the neurovascular unit (NVU), in collaboration with other cells, such as vascular endothelial cells, astrocytes, neurons, and microglia. Microglia, the resident immune cells of the CNS, colonize the brain at embryonic day (E) 9.5 in mice. These cells not only support the development and maturation of neural lineage cells but also help in vascular formation through their extensive migration. Recent studies have demonstrated that pericytes directly contact microglia in the CNS, and their interactions have a profound effect on physiological and pathological aspects. This review summarizes the function of pericytes, focusing on the interplay between pericytes and microglia.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
56
|
Wei Z, Xie Y, Wei M, Zhao H, Ren K, Feng Q, Xu Y. New insights in ferroptosis: Potential therapeutic targets for the treatment of ischemic stroke. Front Pharmacol 2022; 13:1020918. [PMID: 36425577 PMCID: PMC9679292 DOI: 10.3389/fphar.2022.1020918] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/26/2022] [Indexed: 10/22/2023] Open
Abstract
Stroke is a common disease in clinical practice, which seriously endangers people's physical and mental health. The neurovascular unit (NVU) plays a key role in the occurrence and development of ischemic stroke. Different from other classical types of cell death such as apoptosis, necrosis, autophagy, and pyroptosis, ferroptosis is an iron-dependent lipid peroxidation-driven new form of cell death. Interestingly, the function of NVU and stroke development can be regulated by activating or inhibiting ferroptosis. This review systematically describes the NVU in ischemic stroke, provides a comprehensive overview of the regulatory mechanisms and key regulators of ferroptosis, and uncovers the role of ferroptosis in the NVU and the progression of ischemic stroke. We further discuss the latest progress in the intervention of ferroptosis as a therapeutic target for ischemic stroke and summarize the research progress and regulatory mechanism of ferroptosis inhibitors on stroke. In conclusion, ferroptosis, as a new form of cell death, plays a key role in ischemic stroke and is expected to become a new therapeutic target for this disease.
Collapse
Affiliation(s)
- Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingze Wei
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
57
|
Hirunpattarasilp C, Barkaway A, Davis H, Pfeiffer T, Sethi H, Attwell D. Hyperoxia evokes pericyte-mediated capillary constriction. J Cereb Blood Flow Metab 2022; 42:2032-2047. [PMID: 35786054 PMCID: PMC9580167 DOI: 10.1177/0271678x221111598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Oxygen supplementation is regularly prescribed to patients to treat or prevent hypoxia. However, excess oxygenation can lead to reduced cerebral blood flow (CBF) in healthy subjects and worsen the neurological outcome of critically ill patients. Most studies on the vascular effects of hyperoxia focus on arteries but there is no research on the effects on cerebral capillary pericytes, which are major regulators of CBF. Here, we used bright-field imaging of cerebral capillaries and modeling of CBF to show that hyperoxia (95% superfused O2) led to an increase in intracellular calcium level in pericytes and a significant capillary constriction, sufficient to cause an estimated 25% decrease in CBF. Although hyperoxia is reported to cause vascular smooth muscle cell contraction via generation of reactive oxygen species (ROS), endothelin-1 and 20-HETE, we found that increased cytosolic and mitochondrial ROS levels and endothelin release were not involved in the pericyte-mediated capillary constriction. However, a 20-HETE synthesis blocker greatly reduced the hyperoxia-evoked capillary constriction. Our findings establish pericytes as regulators of CBF in hyperoxia and 20-HETE synthesis as an oxygen sensor in CBF regulation. The results also provide a mechanism by which clinically administered oxygen can lead to a worse neurological outcome.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Anna Barkaway
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Harvey Davis
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Thomas Pfeiffer
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - Huma Sethi
- Division of Neurosurgery, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| |
Collapse
|
58
|
Murrant CL, Fletcher NM. Capillary communication: the role of capillaries in sensing the tissue environment, coordinating the microvascular, and controlling blood flow. Am J Physiol Heart Circ Physiol 2022; 323:H1019-H1036. [PMID: 36149771 DOI: 10.1152/ajpheart.00088.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Historically, capillaries have been viewed as the microvascular site for flux of nutrients to cells and removal of waste products. Capillaries are the most numerous blood vessel segment within the tissue, whose vascular wall consists of only a single layer of endothelial cells and are situated within microns of each cell of the tissue, all of which optimizes capillaries for the exchange of nutrients between the blood compartment and the interstitial space of tissues. There is, however, a growing body of evidence to support that capillaries play an important role in sensing the tissue environment, coordinating microvascular network responses, and controlling blood flow. Much of our growing understanding of capillaries stems from work in skeletal muscle and more recent work in the brain, where capillaries can be stimulated by products released from cells of the tissue during increased activity and are able to communicate with upstream and downstream vascular segments, enabling capillaries to sense the activity levels of the tissue and send signals to the microvascular network to coordinate the blood flow response. This review will focus on the emerging role that capillaries play in communication between cells of the tissue and the vascular network required to direct blood flow to active cells in skeletal muscle and the brain. We will also highlight the emerging central role that disruptions in capillary communication may play in blood flow dysregulation, pathophysiology, and disease.
Collapse
Affiliation(s)
- Coral L Murrant
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Nicole M Fletcher
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
59
|
Tomanek RJ. The coronary capillary bed and its role in blood flow and oxygen delivery: A review. Anat Rec (Hoboken) 2022; 305:3199-3211. [PMID: 35521832 PMCID: PMC9796134 DOI: 10.1002/ar.24951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 01/01/2023]
Abstract
The assumption that the coronary capillary blood flow is exclusively regulated by precapillary vessels is not supported by recent data. Rather, the complex coronary capillary bed has unique structural and geometric characteristics that invalidate many assumptions regarding red blood cell (RBC) transport, for example, data based on a single capillary or that increases in flow are the result of capillary recruitment. It is now recognized that all coronary capillaries are open and that their variations in flow are due to structural differences, local O2 demand and delivery, and variations in hematocrit. Recent data reveal that local mechanisms within the capillary bed regulate flow via signaling mechanisms involving RBC signaling and endothelial-associated pericytes that contract and relax in response to humoral and neural signaling. The discovery that pericytes respond to vasoactive signals (e.g., nitric oxide, phenylephrine, and adenosine) underscores the role of these cells in regulating capillary diameter and consequently RBC flux and oxygen delivery. RBCs also affect blood flow by sensing <mml:math xmlns:mml="http://www.w3.org/1998/Math/MathML"><mml:msub><mml:mi>P</mml:mi> <mml:msub><mml:mi>O</mml:mi> <mml:mn>2</mml:mn></mml:msub> </mml:msub> </mml:math> and releasing nitric oxide to facilitate relaxation of pericytes and a consequential capillary dilation. New data indicate that these signaling mechanisms allow control of blood flow in specific coronary capillaries according to their oxygen requirements. In conclusion, mechanisms in the coronary capillary bed facilitate RBC density and transit time, hematocrit, blood flow and O2 delivery, factors that decrease capillary heterogeneity. These findings have important clinical implications for myocardial ischemia and infarction, as well as other vascular diseases.
Collapse
Affiliation(s)
- Robert J. Tomanek
- Department of Anatomy and Cell Biology, Carver College of MedicineUniversity of IowaIowa CityIAUSA
| |
Collapse
|
60
|
Balaratnasingam C, An D, Hein M, Yu P, Yu DY. Studies of the retinal microcirculation using human donor eyes and high-resolution clinical imaging: Insights gained to guide future research in diabetic retinopathy. Prog Retin Eye Res 2022; 94:101134. [PMID: 37154065 DOI: 10.1016/j.preteyeres.2022.101134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The microcirculation plays a key role in delivering oxygen to and removing metabolic wastes from energy-intensive retinal neurons. Microvascular changes are a hallmark feature of diabetic retinopathy (DR), a major cause of irreversible vision loss globally. Early investigators have performed landmark studies characterising the pathologic manifestations of DR. Previous works have collectively informed us of the clinical stages of DR and the retinal manifestations associated with devastating vision loss. Since these reports, major advancements in histologic techniques coupled with three-dimensional image processing has facilitated a deeper understanding of the structural characteristics in the healthy and diseased retinal circulation. Furthermore, breakthroughs in high-resolution retinal imaging have facilitated clinical translation of histologic knowledge to detect and monitor progression of microcirculatory disturbances with greater precision. Isolated perfusion techniques have been applied to human donor eyes to further our understanding of the cytoarchitectural characteristics of the normal human retinal circulation as well as provide novel insights into the pathophysiology of DR. Histology has been used to validate emerging in vivo retinal imaging techniques such as optical coherence tomography angiography. This report provides an overview of our research on the human retinal microcirculation in the context of the current ophthalmic literature. We commence by proposing a standardised histologic lexicon for characterising the human retinal microcirculation and subsequently discuss the pathophysiologic mechanisms underlying key manifestations of DR, with a focus on microaneurysms and retinal ischaemia. The advantages and limitations of current retinal imaging modalities as determined using histologic validation are also presented. We conclude with an overview of the implications of our research and provide a perspective on future directions in DR research.
Collapse
Affiliation(s)
- Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia; Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Australia.
| | - Dong An
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Paula Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| |
Collapse
|
61
|
Smith BC, Tinkey RA, Shaw BC, Williams JL. Targetability of the neurovascular unit in inflammatory diseases of the central nervous system. Immunol Rev 2022; 311:39-49. [PMID: 35909222 PMCID: PMC9489669 DOI: 10.1111/imr.13121] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) is a selectively permeable barrier separating the periphery from the central nervous system (CNS). The BBB restricts the flow of most material into and out of the CNS, including many drugs that could be used as potent therapies. BBB permeability is modulated by several cells that are collectively called the neurovascular unit (NVU). The NVU consists of specialized CNS endothelial cells (ECs), pericytes, astrocytes, microglia, and neurons. CNS ECs maintain a complex "seal" via tight junctions, forming the BBB; breakdown of these tight junctions leads to BBB disruption. Pericytes control the vascular flow within capillaries and help maintain the basal lamina. Astrocytes control much of the flow of material that has moved beyond the CNS EC layer and can form a secondary barrier under inflammatory conditions. Microglia survey the border of the NVU for noxious material. Neuronal activity also plays a role in the maintenance of the BBB. Since astrocytes, pericytes, microglia, and neurons are all able to modulate the permeability of the BBB, understating the complex contributions of each member of the NVU will potentially uncover novel and effective methods for delivery of neurotherapies to the CNS.
Collapse
Affiliation(s)
- Brandon C. Smith
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Department of Biological, Geological, and Environmental SciencesCleveland State UniversityClevelandOhioUSA
| | - Rachel A. Tinkey
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,School of Biomedical SciencesKent State UniversityKentOhioUSA
| | - Benjamin C. Shaw
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Jessica L. Williams
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Brain Health Research Institute, Kent State UniversityKentOhioUSA
| |
Collapse
|
62
|
Deng W, Guo S, van Veluw SJ, Yu Z, Chan SJ, Takase H, Arai K, Ning M, Greenberg SM, Lo EH, Bacskai BJ. Effects of cerebral amyloid angiopathy on the brain vasculome. Aging Cell 2022; 21:e13503. [PMID: 35851991 PMCID: PMC9381891 DOI: 10.1111/acel.13503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 08/27/2021] [Accepted: 10/17/2021] [Indexed: 11/30/2022] Open
Abstract
β‐amyloid (Aβ) deposits in brain blood vessel walls underlie the vascular pathology of Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). Growing evidence has suggested the involvement of cerebrovascular dysfunction in the initiation and progression of cognitive impairment in AD and CAA patients. Therefore, in this study, we assessed the brain vasculome in a mouse model in order to identify cerebrovascular pathways that may be involved in AD and CAA vascular pathogenesis in the context of aging. Brain endothelial cells were isolated from young and old wild‐type mice, and young and old transgenic mice expressing Swedish mutation in amyloid precursor protein and exon 9 deletion in presenilin 1 (APPswe/PSEN1dE9). Microarray profiling of these endothelial transcriptomes demonstrated that accumulation of vascular Aβ in the aging APPswe/PSEN1dE9 mouse is associated with impaired endothelial expression of neurotransmitter receptors and calcium signaling transductors, while the genes involved in cell cycle and inflammation were upregulated. These results suggest that the vascular pathology of AD and CAA may involve the disruption of neurovascular coupling, reactivation of cell cycle in quiescent endothelial cells, and enhanced inflammation. Further dissection of these endothelial mechanisms may offer opportunities to pursue therapies to ameliorate vascular dysfunction in the aging brain of AD and CAA patients.
Collapse
Affiliation(s)
- Wenjun Deng
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
- Department of Neurology Clinical Proteomics Research Center Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Shuzhen Guo
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Susanne J. van Veluw
- Department of Neurology J. Philip Kistler Stroke Research Center Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
- MassGeneral Institute for Neurodegenerative Disease Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Zhanyang Yu
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Su Jing Chan
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Hajime Takase
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - Ken Arai
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| | - MingMing Ning
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
- Department of Neurology Clinical Proteomics Research Center Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Steven M. Greenberg
- Department of Neurology J. Philip Kistler Stroke Research Center Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Eng H. Lo
- Neuroprotection Research Laboratories Department of Radiology and Neurology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
- Department of Neurology Clinical Proteomics Research Center Massachusetts General Hospital Harvard Medical School Boston Massachusetts USA
| | - Brian J. Bacskai
- MassGeneral Institute for Neurodegenerative Disease Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts USA
| |
Collapse
|
63
|
Ioanas HI, Schlegel F, Skachokova Z, Schroeter A, Husak T, Rudin M. Hybrid fiber optic-fMRI for multimodal cell-specific recording and manipulation of neural activity in rodents. NEUROPHOTONICS 2022; 9:032206. [PMID: 35355657 PMCID: PMC8936941 DOI: 10.1117/1.nph.9.3.032206] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/19/2022] [Indexed: 05/08/2023]
Abstract
Significance: Multiscale imaging holds particular relevance to neuroscience, where it helps integrate the cellular and molecular biological scale, which is most accessible to interventions, with holistic organ-level evaluations, most relevant with respect to function. Being inextricably interdisciplinary, multiscale imaging benefits substantially from incremental technology adoption, and a detailed overview of the state-of-the-art is vital to an informed application of imaging methods. Aim: In this article, we lay out the background and methodological aspects of multimodal approaches combining functional magnetic resonance imaging (fMRI) with simultaneous optical measurement or stimulation. Approach: We focus on optical techniques as these allow, in conjunction with genetically encoded proteins (e.g. calcium indicators or optical signal transducers), unprecedented read-out and control specificity for individual cell-types during fMRI experiments, while leveraging non-interfering modalities. Results: A variety of different solutions for optical/fMRI methods has been reported ranging from bulk fluorescence recordings via fiber photometry to high resolution microscopy. In particular, the plethora of optogenetic tools has enabled the transformation of stimulus-evoked fMRI into a cell biological interrogation method. We discuss the capabilities and limitations of these genetically encoded molecular tools in the study of brain phenomena of great methodological and neuropsychiatric interest-such as neurovascular coupling (NVC) and neuronal network mapping. We provide a methodological description of this interdisciplinary field of study, and focus in particular on the limitations of the widely used blood oxygen level dependent (BOLD) signal and how multimodal readouts can shed light on the contributions arising from neurons, astrocytes, or the vasculature. Conclusion: We conclude that information from multiple signaling pathways must be incorporated in future forward models of the BOLD response to prevent erroneous conclusions when using fMRI as a surrogate measure for neural activity. Further, we highlight the potential of direct neuronal stimulation via genetically defined brain networks towards advancing neurophysiological understanding and better estimating effective connectivity.
Collapse
Affiliation(s)
- Horea-Ioan Ioanas
- University of Zurich Institute for Biomedical Engineering, ETH, Zürich, Switzerland
- Massachusetts Institute of Technology, Department of Biological Engineering, Cambridge, Massachusetts, United States
- Dartmouth College, Center for Open Neuroscience, Hanover, New Hampshire, United States
- Address all correspondence to Markus Rudin, ; Horea-Ioan Ioanas,
| | - Felix Schlegel
- University of Zurich Institute for Biomedical Engineering, ETH, Zürich, Switzerland
| | - Zhiva Skachokova
- University of Zurich Institute for Biomedical Engineering, ETH, Zürich, Switzerland
| | - Aileen Schroeter
- University of Zurich Institute for Biomedical Engineering, ETH, Zürich, Switzerland
- University of Zurich, USZ Innovation Hub, Zurich, Switzerland
| | - Tetiana Husak
- Massachusetts Institute of Technology, Department of Electrical Engineering and Computer Science, Cambridge, Massachusetts, United States
| | - Markus Rudin
- University of Zurich Institute for Biomedical Engineering, ETH, Zürich, Switzerland
- The LOOP Zurich, Zurich, Switzerland
- Address all correspondence to Markus Rudin, ; Horea-Ioan Ioanas,
| |
Collapse
|
64
|
Zhang Y, Zhong Y, Zou L, Liu X. Significance of Placental Mesenchymal Stem Cell in Placenta Development and Implications for Preeclampsia. Front Pharmacol 2022; 13:896531. [PMID: 35721156 PMCID: PMC9198303 DOI: 10.3389/fphar.2022.896531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/16/2022] [Indexed: 12/29/2022] Open
Abstract
The well-developed placentation is fundamental for the reproductive pregnancy while the defective placental development is the pathogenetic basis of preeclampsia (PE), a dangerous complication of pregnancy comprising the leading causes of maternal and perinatal morbidity and mortality. Placenta-derived mesenchymal stem cells (PMSCs) are a group of multipotent stem cells that own a potent capacity of differentiating into constitutive cells of vessel walls. Additionally, with the paracrine secretion of various factors, PMSCs inextricably link and interact with other component cells in the placenta, collectively improving the placental vasculature, uterine spiral artery remolding, and uteroplacental interface immunoregulation. Recent studies have further indicated that preeclamptic PMSCs, closely implicated in the abnormal crosstalk between other ambient cells, disturb the homeostasis and development in the placenta. Nevertheless, PMSCs transplantation or PMSCs exosome therapies tend to improve the placental vascular network and trophoblastic functions in the PE model, suggesting PMSCs may be a novel and putative therapeutic strategy for PE. Herein, we provide an overview of the multifaceted contributions of PMSCs in early placental development. Thereinto, the intensive interactions between PMSCs and other component cells in the placenta were particularly highlighted and further extended to the implications in the pathogenesis and therapeutic strategies of PE.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqi Zhong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
65
|
Nakajima T, Nakano S, Kikuchi A, Matsunaga YT. Nailfold capillary patterns correlate with age, gender, lifestyle habits, and fingertip temperature. PLoS One 2022; 17:e0269661. [PMID: 35704663 PMCID: PMC9200324 DOI: 10.1371/journal.pone.0269661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
Nailfold capillaroscopy is a simple and noninvasive imaging tool to visualize the pattern of capillaries. Microvascular abnormalities have been previously observed in autoimmune disease such as systemic sclerosis and diabetes. Thus, early detection of microvascular dysfunction or changes has promising way for the one of the disease preventions. In this study, for routine health checkups, we evaluated the relationship between the structure of nailfold capillaries and lifestyle habits in healthy participants. First, we analyzed the correlation of structural parameters of nailfold capillaries with values of responses to questions on their lifestyle habits in 224 participants. The results suggested that an unhealthy lifestyle, including poor sleeping habits, smoking, intense exercise, and drinking alcohol, causes a change in the pattern of nailfold capillaries. We then investigated whether the pattern of nailfold capillaries changed after a conscious improvement in lifestyle habits. One to two weeks after the self-improvement of lifestyle habits, the hairpin loops sharpened or straightened. In conclusion, this study is the first report indicating a correlation between the structure of nailfold capillaries and lifestyle habits in a non-clinical population. The simple, inexpensive, and noninvasive method using nailfold microscopy can be employed for routine health checkups everywhere even at a bedside.
Collapse
Affiliation(s)
- Tadaaki Nakajima
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- * E-mail: , (TN); (YTM)
| | - Shizuka Nakano
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Department of Materials Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Akihiko Kikuchi
- Department of Materials Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Yukiko T. Matsunaga
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- * E-mail: , (TN); (YTM)
| |
Collapse
|
66
|
Girolamo F, Errede M, Bizzoca A, Virgintino D, Ribatti D. Central Nervous System Pericytes Contribute to Health and Disease. Cells 2022; 11:1707. [PMID: 35626743 PMCID: PMC9139243 DOI: 10.3390/cells11101707] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/11/2022] Open
Abstract
Successful neuroprotection is only possible with contemporary microvascular protection. The prevention of disease-induced vascular modifications that accelerate brain damage remains largely elusive. An improved understanding of pericyte (PC) signalling could provide important insight into the function of the neurovascular unit (NVU), and into the injury-provoked responses that modify cell-cell interactions and crosstalk. Due to sharing the same basement membrane with endothelial cells, PCs have a crucial role in the control of endothelial, astrocyte, and oligodendrocyte precursor functions and hence blood-brain barrier stability. Both cerebrovascular and neurodegenerative diseases impair oxygen delivery and functionally impair the NVU. In this review, the role of PCs in central nervous system health and disease is discussed, considering their origin, multipotency, functions and also dysfunction, focusing on new possible avenues to modulate neuroprotection. Dysfunctional PC signalling could also be considered as a potential biomarker of NVU pathology, allowing us to individualize therapeutic interventions, monitor responses, or predict outcomes.
Collapse
Affiliation(s)
- Francesco Girolamo
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Mariella Errede
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Antonella Bizzoca
- Physiology Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Daniela Virgintino
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Domenico Ribatti
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| |
Collapse
|
67
|
Korte N, Ilkan Z, Pearson CL, Pfeiffer T, Singhal P, Rock JR, Sethi H, Gill D, Attwell D, Tammaro P. The Ca2+-gated channel TMEM16A amplifies capillary pericyte contraction and reduces cerebral blood flow after ischemia. J Clin Invest 2022; 132:e154118. [PMID: 35316222 PMCID: PMC9057602 DOI: 10.1172/jci154118] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/16/2022] [Indexed: 11/26/2022] Open
Abstract
Pericyte-mediated capillary constriction decreases cerebral blood flow in stroke after an occluded artery is unblocked. The determinants of pericyte tone are poorly understood. We show that a small rise in cytoplasmic Ca2+ concentration ([Ca2+]i) in pericytes activated chloride efflux through the Ca2+-gated anion channel TMEM16A, thus depolarizing the cell and opening voltage-gated calcium channels. This mechanism strongly amplified the pericyte [Ca2+]i rise and capillary constriction evoked by contractile agonists and ischemia. In a rodent stroke model, TMEM16A inhibition slowed the ischemia-evoked pericyte [Ca2+]i rise, capillary constriction, and pericyte death; reduced neutrophil stalling; and improved cerebrovascular reperfusion. Genetic analysis implicated altered TMEM16A expression in poor patient recovery from ischemic stroke. Thus, pericyte TMEM16A is a crucial regulator of cerebral capillary function and a potential therapeutic target for stroke and possibly other disorders of impaired microvascular flow, such as Alzheimer's disease and vascular dementia.
Collapse
Affiliation(s)
- Nils Korte
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Zeki Ilkan
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Claire L. Pearson
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Thomas Pfeiffer
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Prabhav Singhal
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Jason R. Rock
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Huma Sethi
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, St Mary’s Hospital, Imperial College London, London, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Paolo Tammaro
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
68
|
Roy TK, Secomb TW. Functional implications of microvascular heterogeneity for oxygen uptake and utilization. Physiol Rep 2022; 10:e15303. [PMID: 35581743 PMCID: PMC9114652 DOI: 10.14814/phy2.15303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023] Open
Abstract
In the vascular system, an extensive network structure provides convective and diffusive transport of oxygen to tissue. In the microcirculation, parameters describing network structure, blood flow, and oxygen transport are highly heterogeneous. This heterogeneity can strongly affect oxygen supply and organ function, including reduced oxygen uptake in the lung and decreased oxygen delivery to tissue. The causes of heterogeneity can be classified as extrinsic or intrinsic. Extrinsic heterogeneity refers to variations in oxygen demand in the systemic circulation or oxygen supply in the lungs. Intrinsic heterogeneity refers to structural heterogeneity due to stochastic growth of blood vessels and variability in flow pathways due to geometric constraints, and resulting variations in blood flow and hematocrit. Mechanisms have evolved to compensate for heterogeneity and thereby improve oxygen uptake in the lung and delivery to tissue. These mechanisms, which involve long-term structural adaptation and short-term flow regulation, depend on upstream responses conducted along vessel walls, and work to redistribute flow and maintain blood and tissue oxygenation. Mathematically, the variance of a functional quantity such as oxygen delivery that depends on two or more heterogeneous variables can be reduced if one of the underlying variables is controlled by an appropriate compensatory mechanism. Ineffective regulatory mechanisms can result in poor oxygen delivery even in the presence of adequate overall tissue perfusion. Restoration of endothelial function, and specifically conducted responses, should be considered when addressing tissue hypoxemia and organ failure in clinical settings.
Collapse
Affiliation(s)
- Tuhin K. Roy
- Department of AnesthesiologyMayo ClinicRochesterMinnesotaUSA
| | | |
Collapse
|
69
|
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS 2022; 19:29. [PMID: 35410231 PMCID: PMC8996682 DOI: 10.1186/s12987-022-00312-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The response of the blood-brain barrier (BBB) following a stroke, including subarachnoid hemorrhage (SAH), has been studied extensively. The main components of this reaction are endothelial cells, pericytes, and astrocytes that affect microglia, neurons, and vascular smooth muscle cells. SAH induces alterations in individual BBB cells, leading to brain homeostasis disruption. Recent experiments have uncovered many pathophysiological cascades affecting the BBB following SAH. Targeting some of these pathways is important for restoring brain function following SAH. BBB injury occurs immediately after SAH and has long-lasting consequences, but most changes in the pathophysiological cascades occur in the first few days following SAH. These changes determine the development of early brain injury as well as delayed cerebral ischemia. SAH-induced neuroprotection also plays an important role and weakens the negative impact of SAH. Supporting some of these beneficial cascades while attenuating the major pathophysiological pathways might be decisive in inhibiting the negative impact of bleeding in the subarachnoid space. In this review, we attempt a comprehensive overview of the current knowledge on the molecular and cellular changes in the BBB following SAH and their possible modulation by various drugs and substances.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Klaudia Lakatosová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
70
|
Wang X, Li J, Bian Y, Zhao C, Li J, Li X. pH regulates the lumen diameter of tissue-engineered capillaries. Exp Ther Med 2022; 23:284. [PMID: 35317437 PMCID: PMC8908470 DOI: 10.3892/etm.2022.11212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/03/2021] [Indexed: 11/24/2022] Open
Abstract
Angiogenesis is vital in tissue engineering and the size of the capillary lumen diameter directly affects vascular function. Therefore, the involvement of the pH in the regulation of the capillary lumen diameter was investigated in the present study. The cytosolic pH of different pH medium groups was measured using flow cytometry. Bromodeoxyuridine staining and wound-healing assays were performed to detect cell proliferation and migration, respectively. The expression of angiogenesis-related genes was detected using reverse transcription-quantitative PCR. In addition, cell tube formation under different pH conditions was assessed using a tube formation assay and a 3D Matrigel® model. The results indicated that a change in the pH value of the culture medium affected the cytosolic pH of the endothelial cells, which then led to a change in vascular diameter. When the medium's pH ranged from 7.4 to 7.6, the diameter of the lumen formed in the Matrigel was suitable for capillary formation in tissue engineering. The present results revealed an important role for the pH in the process of capillary formation and provided insight for pH regulation during endothelial cell tube formation and angiogenesis in tissue engineering.
Collapse
Affiliation(s)
- Xiaolin Wang
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jing Li
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yongqian Bian
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Congying Zhao
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jinqing Li
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xueyong Li
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
71
|
Dion-Albert L, Bandeira Binder L, Daigle B, Hong-Minh A, Lebel M, Menard C. Sex differences in the blood-brain barrier: Implications for mental health. Front Neuroendocrinol 2022; 65:100989. [PMID: 35271863 DOI: 10.1016/j.yfrne.2022.100989] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/07/2022] [Accepted: 02/19/2022] [Indexed: 12/13/2022]
Abstract
Prevalence of mental disorders, including major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia (SZ) are increasing at alarming rates in our societies. Growing evidence points toward major sex differences in these conditions, and high rates of treatment resistance support the need to consider novel biological mechanisms outside of neuronal function to gain mechanistic insights that could lead to innovative therapies. Blood-brain barrier alterations have been reported in MDD, BD and SZ. Here, we provide an overview of sex-specific immune, endocrine, vascular and transcriptional-mediated changes that could affect neurovascular integrity and possibly contribute to the pathogenesis of mental disorders. We also identify pitfalls in current literature and highlight promising vascular biomarkers. Better understanding of how these adaptations can contribute to mental health status is essential not only in the context of MDD, BD and SZ but also cardiovascular diseases and stroke which are associated with higher prevalence of these conditions.
Collapse
Affiliation(s)
- Laurence Dion-Albert
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Luisa Bandeira Binder
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Beatrice Daigle
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Amandine Hong-Minh
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - Manon Lebel
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Caroline Menard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada.
| |
Collapse
|
72
|
Wareham LK, Liddelow SA, Temple S, Benowitz LI, Di Polo A, Wellington C, Goldberg JL, He Z, Duan X, Bu G, Davis AA, Shekhar K, Torre AL, Chan DC, Canto-Soler MV, Flanagan JG, Subramanian P, Rossi S, Brunner T, Bovenkamp DE, Calkins DJ. Solving neurodegeneration: common mechanisms and strategies for new treatments. Mol Neurodegener 2022; 17:23. [PMID: 35313950 PMCID: PMC8935795 DOI: 10.1186/s13024-022-00524-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Across neurodegenerative diseases, common mechanisms may reveal novel therapeutic targets based on neuronal protection, repair, or regeneration, independent of etiology or site of disease pathology. To address these mechanisms and discuss emerging treatments, in April, 2021, Glaucoma Research Foundation, BrightFocus Foundation, and the Melza M. and Frank Theodore Barr Foundation collaborated to bring together key opinion leaders and experts in the field of neurodegenerative disease for a virtual meeting titled "Solving Neurodegeneration". This "think-tank" style meeting focused on uncovering common mechanistic roots of neurodegenerative disease and promising targets for new treatments, catalyzed by the goal of finding new treatments for glaucoma, the world's leading cause of irreversible blindness and the common interest of the three hosting foundations. Glaucoma, which causes vision loss through degeneration of the optic nerve, likely shares early cellular and molecular events with other neurodegenerative diseases of the central nervous system. Here we discuss major areas of mechanistic overlap between neurodegenerative diseases of the central nervous system: neuroinflammation, bioenergetics and metabolism, genetic contributions, and neurovascular interactions. We summarize important discussion points with emphasis on the research areas that are most innovative and promising in the treatment of neurodegeneration yet require further development. The research that is highlighted provides unique opportunities for collaboration that will lead to efforts in preventing neurodegeneration and ultimately vision loss.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Sally Temple
- Neural Stem Cell Institute, NY, 12144, Rensselaer, USA
| | - Larry I Benowitz
- Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, CA, Palo Alto, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, MA, Boston, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Albert A Davis
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, CA, 91125, Pasadena, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Aurora, CO, USA
| | - John G Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | | | | | | | | | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
73
|
Gagliano G, Monteverdi A, Casali S, Laforenza U, Gandini Wheeler-Kingshott CAM, D’Angelo E, Mapelli L. Non-Linear Frequency Dependence of Neurovascular Coupling in the Cerebellar Cortex Implies Vasodilation-Vasoconstriction Competition. Cells 2022; 11:1047. [PMID: 35326498 PMCID: PMC8947624 DOI: 10.3390/cells11061047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 01/28/2023] Open
Abstract
Neurovascular coupling (NVC) is the process associating local cerebral blood flow (CBF) to neuronal activity (NA). Although NVC provides the basis for the blood oxygen level dependent (BOLD) effect used in functional MRI (fMRI), the relationship between NVC and NA is still unclear. Since recent studies reported cerebellar non-linearities in BOLD signals during motor tasks execution, we investigated the NVC/NA relationship using a range of input frequencies in acute mouse cerebellar slices of vermis and hemisphere. The capillary diameter increased in response to mossy fiber activation in the 6-300 Hz range, with a marked inflection around 50 Hz (vermis) and 100 Hz (hemisphere). The corresponding NA was recorded using high-density multi-electrode arrays and correlated to capillary dynamics through a computational model dissecting the main components of granular layer activity. Here, NVC is known to involve a balance between the NMDAR-NO pathway driving vasodilation and the mGluRs-20HETE pathway driving vasoconstriction. Simulations showed that the NMDAR-mediated component of NA was sufficient to explain the time course of the capillary dilation but not its non-linear frequency dependence, suggesting that the mGluRs-20HETE pathway plays a role at intermediate frequencies. These parallel control pathways imply a vasodilation-vasoconstriction competition hypothesis that could adapt local hemodynamics at the microscale bearing implications for fMRI signals interpretation.
Collapse
Affiliation(s)
- Giuseppe Gagliano
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
| | - Anita Monteverdi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Stefano Casali
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
| | - Umberto Laforenza
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Claudia A. M. Gandini Wheeler-Kingshott
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London WC1N3 BG, UK
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
| |
Collapse
|
74
|
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer's disease: New perspectives. Brain Pathol 2022; 32:e13061. [PMID: 35289012 PMCID: PMC9616094 DOI: 10.1111/bpa.13061] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Collapse
Affiliation(s)
- Robert A Fisher
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - Seth Love
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| |
Collapse
|
75
|
Császár E, Lénárt N, Cserép C, Környei Z, Fekete R, Pósfai B, Balázsfi D, Hangya B, Schwarcz AD, Szabadits E, Szöllősi D, Szigeti K, Máthé D, West BL, Sviatkó K, Brás AR, Mariani JC, Kliewer A, Lenkei Z, Hricisák L, Benyó Z, Baranyi M, Sperlágh B, Menyhárt Á, Farkas E, Dénes Á. Microglia modulate blood flow, neurovascular coupling, and hypoperfusion via purinergic actions. J Exp Med 2022; 219:e20211071. [PMID: 35201268 PMCID: PMC8932534 DOI: 10.1084/jem.20211071] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/28/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022] Open
Abstract
Microglia, the main immunocompetent cells of the brain, regulate neuronal function, but their contribution to cerebral blood flow (CBF) regulation has remained elusive. Here, we identify microglia as important modulators of CBF both under physiological conditions and during hypoperfusion. Microglia establish direct, dynamic purinergic contacts with cells in the neurovascular unit that shape CBF in both mice and humans. Surprisingly, the absence of microglia or blockade of microglial P2Y12 receptor (P2Y12R) substantially impairs neurovascular coupling in mice, which is reiterated by chemogenetically induced microglial dysfunction associated with impaired ATP sensitivity. Hypercapnia induces rapid microglial calcium changes, P2Y12R-mediated formation of perivascular phylopodia, and microglial adenosine production, while depletion of microglia reduces brain pH and impairs hypercapnia-induced vasodilation. Microglial actions modulate vascular cyclic GMP levels but are partially independent of nitric oxide. Finally, microglial dysfunction markedly impairs P2Y12R-mediated cerebrovascular adaptation to common carotid artery occlusion resulting in hypoperfusion. Thus, our data reveal a previously unrecognized role for microglia in CBF regulation, with broad implications for common neurological diseases.
Collapse
Affiliation(s)
- Eszter Császár
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Nikolett Lénárt
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zsuzsanna Környei
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Rebeka Fekete
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Diána Balázsfi
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Hangya
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Anett D. Schwarcz
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Szabadits
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Dávid Szöllősi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
| | | | - Katalin Sviatkó
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Ana Rita Brás
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Jean-Charles Mariani
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, Paris, France
| | - Andrea Kliewer
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, Paris, France
| | - Zsolt Lenkei
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, Paris, France
| | - László Hricisák
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Mária Baranyi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine, University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine, University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ádám Dénes
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
76
|
Jackson JG, Krizman E, Takano H, Lee M, Choi GH, Putt ME, Robinson MB. Activation of Glutamate Transport Increases Arteriole Diameter in v ivo: Implications for Neurovascular Coupling. Front Cell Neurosci 2022; 16:831061. [PMID: 35308116 PMCID: PMC8930833 DOI: 10.3389/fncel.2022.831061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 11/21/2022] Open
Abstract
In order to meet the energetic demands of cell-to-cell signaling, increases in local neuronal signaling are matched by a coordinated increase in local blood flow, termed neurovascular coupling. Multiple different signals from neurons, astrocytes, and pericytes contribute to this control of blood flow. Previously, several groups demonstrated that inhibition/ablation of glutamate transporters attenuates the neurovascular response. However, it was not determined if glutamate transporter activation was sufficient to increase blood flow. Here, we used multiphoton imaging to monitor the diameter of fluorescently labeled cortical arterioles in anesthetized C57/B6J mice. We delivered vehicle, glutamate transporter substrates, or a combination of a glutamate transporter substrate with various pharmacologic agents via a glass micropipette while simultaneously visualizing changes in arteriole diameter. We developed a novel image analysis method to automate the measurement of arteriole diameter in these time-lapse analyses. Using this workflow, we first conducted pilot experiments in which we focally applied L-glutamate, D-aspartate, or L-threo-hydroxyaspartate (L-THA) and measured arteriole responses as proof of concept. We subsequently applied the selective glutamate transport substrate L-THA (applied at concentrations that do not activate glutamate receptors). We found that L-THA evoked a significantly larger dilation than that observed with focal saline application. This response was blocked by co-application of the potent glutamate transport inhibitor, L-(2S,3S)-3-[3-[4-(trifluoromethyl)-benzoylamino]benzyloxy]-aspartate (TFB-TBOA). Conversely, we were unable to demonstrate a reduction of this effect through co-application of a cocktail of glutamate and GABA receptor antagonists. These studies provide the first direct evidence that activation of glutamate transport is sufficient to increase arteriole diameter. We explored potential downstream mechanisms mediating this transporter-mediated dilation by using a Ca2+ chelator or inhibitors of reversed-mode Na+/Ca2+ exchange, nitric oxide synthetase, or cyclo-oxygenase. The estimated effects and confidence intervals suggested some form of inhibition for a number of these inhibitors. Limitations to our study design prevented definitive conclusions with respect to these downstream inhibitors; these limitations are discussed along with possible next steps. Understanding the mechanisms that control blood flow are important because changes in blood flow/energy supply are implicated in several neurodegenerative disorders and are used as a surrogate measure of neuronal activity in widely used techniques such as functional magnetic resonance imaging (fMRI).
Collapse
Affiliation(s)
- Joshua G. Jackson
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, United States
| | - Elizabeth Krizman
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, United States
| | - Hajime Takano
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - Meredith Lee
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Grace H. Choi
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Mary E. Putt
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael B. Robinson
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
77
|
Shaw K, Boyd K, Anderle S, Hammond-Haley M, Amin D, Bonnar O, Hall CN. Gradual Not Sudden Change: Multiple Sites of Functional Transition Across the Microvascular Bed. Front Aging Neurosci 2022; 13:779823. [PMID: 35237142 PMCID: PMC8885127 DOI: 10.3389/fnagi.2021.779823] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/20/2021] [Indexed: 01/03/2023] Open
Abstract
In understanding the role of the neurovascular unit as both a biomarker and target for disease interventions, it is vital to appreciate how the function of different components of this unit change along the vascular tree. The cells of the neurovascular unit together perform an array of vital functions, protecting the brain from circulating toxins and infection, while providing nutrients and clearing away waste products. To do so, the brain's microvasculature dilates to direct energy substrates to active neurons, regulates access to circulating immune cells, and promotes angiogenesis in response to decreased blood supply, as well as pulsating to help clear waste products and maintain the oxygen supply. Different parts of the cerebrovascular tree contribute differently to various aspects of these functions, and previously, it has been assumed that there are discrete types of vessel along the vascular network that mediate different functions. Another option, however, is that the multiple transitions in function that occur across the vascular network do so at many locations, such that vascular function changes gradually, rather than in sharp steps between clearly distinct vessel types. Here, by reference to new data as well as by reviewing historical and recent literature, we argue that this latter scenario is likely the case and that vascular function gradually changes across the network without clear transition points between arteriole, precapillary arteriole and capillary. This is because classically localized functions are in fact performed by wide swathes of the vasculature, and different functional markers start and stop being expressed at different points along the vascular tree. Furthermore, vascular branch points show alterations in their mural cell morphology that suggest functional specializations irrespective of their position within the network. Together this work emphasizes the need for studies to consider where transitions of different functions occur, and the importance of defining these locations, in order to better understand the vascular network and how to target it to treat disease.
Collapse
Affiliation(s)
- Kira Shaw
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Katie Boyd
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Silvia Anderle
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | | | - Davina Amin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Orla Bonnar
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown Navy Yard, MA, United States
| | - Catherine N. Hall
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| |
Collapse
|
78
|
Hartmann DA, Coelho-Santos V, Shih AY. Pericyte Control of Blood Flow Across Microvascular Zones in the Central Nervous System. Annu Rev Physiol 2022; 84:331-354. [PMID: 34672718 PMCID: PMC10480047 DOI: 10.1146/annurev-physiol-061121-040127] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The vast majority of the brain's vascular length is composed of capillaries, where our understanding of blood flow control remains incomplete. This review synthesizes current knowledge on the control of blood flow across microvascular zones by addressing issues with nomenclature and drawing on new developments from in vivo optical imaging and single-cell transcriptomics. Recent studies have highlighted important distinctions in mural cell morphology, gene expression, and contractile dynamics, which can explain observed differences in response to vasoactive mediators between arteriole, transitional, and capillary zones. Smooth muscle cells of arterioles and ensheathing pericytes of the arteriole-capillary transitional zone control large-scale, rapid changes in blood flow. In contrast, capillary pericytes downstream of the transitional zone act on slower and smaller scales and are involved in establishing resting capillary tone and flow heterogeneity. Many unresolved issues remain, including the vasoactive mediators that activate the different pericyte types in vivo, the role of pericyte-endothelial communication in conducting signals from capillaries to arterioles, and how neurological disease affects these mechanisms.
Collapse
Affiliation(s)
- David A Hartmann
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Vanessa Coelho-Santos
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA;
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA;
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
79
|
Jacobsen NL, Norton CE, Shaw RL, Cornelison DDW, Segal SS. Myofibre injury induces capillary disruption and regeneration of disorganized microvascular networks. J Physiol 2022; 600:41-60. [PMID: 34761825 PMCID: PMC8965732 DOI: 10.1113/jp282292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023] Open
Abstract
Injury to skeletal muscle disrupts myofibres and their microvascular supply. While the regeneration of myofibres is well described, little is known of how the microcirculation is affected by skeletal muscle injury or its recovery during regeneration. Nevertheless, the microvasculature must also recover to restore skeletal muscle function. We aimed to define the nature of microvascular damage and time course of repair during muscle injury and regeneration induced by the myotoxin BaCl2 . To test the hypothesis that microvascular disruption occurred secondary to myofibre injury, isolated microvessels were exposed to BaCl2 or the myotoxin was injected into the gluteus maximus (GM) muscle of mice. In isolated microvessels, BaCl2 depolarized smooth muscle cells (SMCs) and endothelial cells while increasing intracellular calcium in SMCs but did not elicit death of either cell type. At 1 day post-injury (dpi) of the GM, capillary fragmentation coincided with myofibre degeneration while arteriolar and venular networks remained intact; neutrophil depletion before injury did not prevent capillary damage. Perfused capillary networks reformed by 5 dpi in association with more terminal arterioles and were dilated through 10 dpi. With no change in microvascular area or branch point number in regenerating capillary networks, fewer capillaries aligned with myofibres and were no longer organized into microvascular units. By 21 dpi, capillary orientation and microvascular unit organization were no longer different from uninjured GM. We conclude that following their disruption secondary to myofibre damage, capillaries regenerate as disorganized networks that remodel into microvascular units as regenerated myofibres mature. KEY POINTS: Skeletal muscle regenerates after injury; however, the nature of microvascular damage and repair is poorly understood. Here, the myotoxin BaCl2 , a standard experimental method of acute skeletal muscle injury, was used to investigate the response of the microcirculation to local injury of intact muscle. Intramuscular injection of BaCl2 induced capillary fragmentation with myofibre degeneration; arteriolar and venular networks remained intact. Direct exposure to BaCl2 did not kill microvascular endothelial cells or smooth muscle cells. Dilated capillary networks reformed by 5 days post-injury (dpi) in association with more terminal arterioles. Capillary orientation remained disorganized through 10 dpi. Capillaries realigned with myofibres and reorganized into microvascular units by 21 dpi, which coincides with the recovery of vasomotor control and maturation of nascent myofibres. Skeletal muscle injury disrupts its capillary supply secondary to myofibre degeneration. Reorganization of regenerating microvascular networks accompanies the recovery of blood flow regulation.
Collapse
Affiliation(s)
- Nicole L. Jacobsen
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Charles E. Norton
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Rebecca L. Shaw
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - D. D. W. Cornelison
- Biological Sciences, University of Missouri, Columbia, MO, USA,Christopher S. Bond Life Sciences Center, University of MO, Columbia, MO, USA
| | - Steven S. Segal
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
80
|
Winkelman MA, Kim DY, Kakarla S, Grath A, Silvia N, Dai G. Interstitial flow enhances the formation, connectivity, and function of 3D brain microvascular networks generated within a microfluidic device. LAB ON A CHIP 2021; 22:170-192. [PMID: 34881385 PMCID: PMC9257897 DOI: 10.1039/d1lc00605c] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The bulk flow of interstitial fluid through tissue is an important factor in human biology, including the development of brain microvascular networks (MVNs) with the blood-brain barrier (BBB). Bioengineering perfused, functional brain MVNs has great potential for modeling neurovascular diseases and drug delivery. However, most in vitro models of brain MVNs do not implement interstitial flow during the generation of microvessels. Using a microfluidic device (MFD), we cultured primary human brain endothelial cells (BECs), pericytes, and astrocytes within a 3D fibrin matrix with (flow) and without (static) interstitial flow. We found that the bulk flow of interstitial fluid was beneficial for both BEC angiogenesis and vasculogenesis. Brain MVNs cultured under flow conditions achieved anastomosis and were perfusable, whereas those under static conditions lacked connectivity and the ability to be perfused. Compared to static culture, microvessels developed in flow culture exhibited an enhanced vessel area, branch length and diameter, connectivity, and longevity. Although there was no change in pericyte coverage of microvessels, a slight increase in astrocyte coverage was observed under flow conditions. In addition, the immunofluorescence intensity of basal lamina proteins, collagen IV and laminin, was nearly doubled in flow culture. Lastly, the barrier function of brain microvessels was enhanced under flow conditions, as demonstrated by decreased dextran permeability. Taken together, these results highlighted the importance of interstitial flow in the in vitro generation of perfused brain MVNs with characteristics similar to those of the human BBB.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Diana Y Kim
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Shravani Kakarla
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Alexander Grath
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Nathaniel Silvia
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, 805 Columbus Ave, ISEC 224, Boston, MA, 02115, USA.
| |
Collapse
|
81
|
Xu H, Zong Y, Yu J, Jiang C, Zhu H, Sun X. Retinal Microvascular Reactivity in Chronic Cigarette Smokers and Non-smokers: An Observational Cross-Sectional Study. Front Med (Lausanne) 2021; 8:782010. [PMID: 34988096 PMCID: PMC8720846 DOI: 10.3389/fmed.2021.782010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Purpose: To evaluate the changes in the retinal microvasculature and its reactivity in chronic cigarette smokers. Methods: Thirty-four male chronic cigarette smokers and 18 male non-smokers were enrolled. Optical coherence tomography angiography was used to measure the perfused retinal vessel densities (PVDs) of the peripapillary and parafoveal areas at baseline and during phase IV of the Valsalva maneuver (VM-IV). Systemic blood pressure and intraocular pressure were also measured. Results: The baseline PVD in the peripapillary area of the smokers was significantly lower than the non-smokers (59.56 ± 2.26% vs. 61.67 ± 3.58%, respectively; P = 0.005). However, there was no significant difference in the foveal avascular zone or parafoveal PVD between the two groups. During VM-IV, the peripapillary PVD of the smokers decreased by 1.13 ± 3.50%, which was significantly less than that of the non-smokers (−3.83 ± 4.26%, P < 0.05). Similarly, the parafoveal PVD of the smokers decreased by 5.49 ± 9.70%, which was significantly less than the percentage change of the non-smokers (−13.01 ± 8.39%, P < 0.05). There was no significant difference in the percentage change in systemic blood pressure parameters between the two groups. Conclusion: The retinal microvasculature and its reactivity were impaired in chronic smokers compared with non-smokers. The extent of impairment differed among different regions of the fundus.
Collapse
Affiliation(s)
- Huan Xu
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China
- Key Laboratory of Myopia of State Health Ministry and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Yuan Zong
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China
- Key Laboratory of Myopia of State Health Ministry and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Jian Yu
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China
- Key Laboratory of Myopia of State Health Ministry and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| | - Chunhui Jiang
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China
- Key Laboratory of Myopia of State Health Ministry and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
- *Correspondence: Chunhui Jiang
| | - Haohao Zhu
- Department of Ophthalmology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
- Haohao Zhu
| | - Xinghuai Sun
- Department of Ophthalmology and Visual Science, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College of Fudan University, Shanghai, China
- Key Laboratory of Myopia of State Health Ministry and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai, China
| |
Collapse
|
82
|
Shao X, Guo F, Shou Q, Wang K, Jann K, Yan L, Toga AW, Zhang P, Wang DJJ. Laminar perfusion imaging with zoomed arterial spin labeling at 7 Tesla. Neuroimage 2021; 245:118724. [PMID: 34780918 PMCID: PMC8727512 DOI: 10.1016/j.neuroimage.2021.118724] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/23/2021] [Accepted: 11/11/2021] [Indexed: 10/19/2022] Open
Abstract
Laminar fMRI based on BOLD and CBV contrast at ultrahigh magnetic fields has been applied for studying the dynamics of mesoscopic brain networks. However, the quantitative interpretations of BOLD/CBV fMRI results are confounded by different baseline physiology across cortical layers. Here we introduce a novel 3D zoomed pseudo-continuous arterial spin labeling (pCASL) technique at 7T that offers the capability for quantitative measurements of laminar cerebral blood flow (CBF) both at rest and during task activation with high spatial specificity and sensitivity. We found arterial transit time in superficial layers is ∼100 ms shorter than in middle/deep layers revealing the time course of labeled blood flowing from pial arteries to downstream microvasculature. Resting state CBF peaked in the middle layers which is highly consistent with microvascular density measured from human cortex specimens. Finger tapping induced a robust two-peak laminar profile of CBF increases in the superficial (somatosensory and premotor input) and deep (spinal output) layers of M1, while finger brushing task induced a weaker CBF increase in superficial layers (somatosensory input). This observation is highly consistent with reported laminar profiles of CBV activation on M1. We further demonstrated that visuospatial attention induced a predominant CBF increase in deep layers and a smaller CBF increase on top of the lower baseline CBF in superficial layers of V1 (feedback cortical input), while stimulus driven activity peaked in the middle layers (feedforward thalamic input). With the capability for quantitative CBF measurements both at baseline and during task activation, high-resolution ASL perfusion fMRI at 7T provides an important tool for in vivo assessment of neurovascular function and metabolic activities of neural circuits across cortical layers.
Collapse
Affiliation(s)
- Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave, Los Angeles, CA 90033, USA
| | - Fanhua Guo
- State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qinyang Shou
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave, Los Angeles, CA 90033, USA
| | - Kai Wang
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave, Los Angeles, CA 90033, USA
| | - Kay Jann
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave, Los Angeles, CA 90033, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Lirong Yan
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave, Los Angeles, CA 90033, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Peng Zhang
- State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave, Los Angeles, CA 90033, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
83
|
Hudson N, Campbell M. Tight Junctions of the Neurovascular Unit. Front Mol Neurosci 2021; 14:752781. [PMID: 34867185 PMCID: PMC8640090 DOI: 10.3389/fnmol.2021.752781] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
The homeostatic balance of the brain and retina is maintained by the presence of the blood-brain and inner blood-retinal barrier (BBB/iBRB, respectively) which are highly specialized barriers. Endothelial cells forming the lining of these blood vessels are interconnected by the presence of tight junctions which form the BBB and iBRB. These tight junctions, formed of numerous interacting proteins, enable the entry of molecules into neural tissues while restricting the entry of harmful material such as anaphylatoxins, bacteria and viruses. If the tight junction complex becomes dysregulated due to changes in expression levels of one or more of the components, this can have detrimental effects leading to brain and retinal pathology.
Collapse
Affiliation(s)
- Natalie Hudson
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, Ireland
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, Ireland
| |
Collapse
|
84
|
Ternifi R, Wang Y, Polley EC, Fazzio RT, Fatemi M, Alizad A. Quantitative Biomarkers for Cancer Detection Using Contrast-Free Ultrasound High-Definition Microvessel Imaging: Fractal Dimension, Murray's Deviation, Bifurcation Angle & Spatial Vascularity Pattern. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:3891-3900. [PMID: 34329160 PMCID: PMC8668387 DOI: 10.1109/tmi.2021.3101669] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
A growing body of evidence indicates that there is a strong correlation between microvascular morphological features and malignant tumors. Therefore, quantification of these features might allow more accurate differentiation of benign and malignant tumors. The main objective of this research project is to improve the quantification of microvascular networks depicted in contrast-free ultrasound microvessel images. To achieve this goal, a new series of quantitative microvessel morphological parameters are introduced for differentiation of breast masses using contrast-free ultrasound-based high-definition microvessel imaging (HDMI). Using HDMI, we quantified and analyzed four new parameters: 1) microvessel fractal dimension (mvFD), a marker of tumor microvascular complexity; 2) Murray's deviation (MD), the diameter mismatch, defined as the deviation from Murray's law; 3) bifurcation angle (BA), abnormally decreased angle; and 4) spatial vascular pattern (SVP), indicating tumor vascular distribution pattern, either intratumoral or peritumoral. The new biomarkers have been tested on 60 patients with breast masses. Validation of the feature's extraction algorithm was performed using a synthetic data set. All the proposed parameters had the power to discriminate the breast lesion malignancy (p < 0.05), displaying BA as the most sensitive test, with a sensitivity of 90.6%, and mvFD as the most specific test, with a specificity of 92%. The results of all four new biomarkers showed an AUC = 0.889, sensitivity of 80% and specificity of 91.4% In conclusion, the added value of the proposed quantitative morphological parameters, as new biomarkers of angiogenesis within breast masses, paves the way for more accurate breast cancer detection with higher specificity.
Collapse
|
85
|
Fremont-Smith M, Gherlone N, Smith N, Tisdall P, Ricke DO. Models for COVID-19 Early Cardiac Pathology Following SARS-CoV-2 Infection. Int J Infect Dis 2021; 113:331-335. [PMID: 34592443 PMCID: PMC8473263 DOI: 10.1016/j.ijid.2021.09.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES The clinical manifestations of COVID-19 associated cardiac complications are heterogeneous, ranging from asymptomatic to severe symptoms, including arrhythmias and cardiogenic shock. For COVID-19 patients with cardiac sequela, only a small subset of patients have myocarditis; the pathogenesis of cardiac sequela caused by SARS-CoV-2 other than microthrombi associated sequela remains to be determined. METHODS Retrospective analysis of 71 heart autopsy specimens from COVID-19 and putative COVID-19 in the NIH COVID Digital Pathology Repository. RESULTS The most consistent observation was localized myocardial cell death not associated with either myocarditis or microthrombi. Red blood cells were typically absent from capillaries but, when observed, were predominately in linear clusters (stacks) of adjacent cells. CONCLUSIONS Based on our retrospective analysis, we propose that localized ischemia and subsequent cell death by anoxia contributes to the cardiac pathogenesis in some COVID-19 patients. We propose two new models predicting vasoconstriction of cardiac pericyte cells induced by elevated histamine from hyper-activated mast cells or direct infection. We propose that impeded blood flow and cell death by anoxia are initial steps in the development of SARS-CoV-2 induced cardiac injury in COVID-19 patients independent of microthrombi or myocarditis.
Collapse
Affiliation(s)
| | - Nicole Gherlone
- AMITA Health Saint Joseph Hospital Chicago, Chicago, IL 60657, USA.
| | - Nora Smith
- Massachusetts Institute of Technology, Lincoln Laboratory, Lexington, MA, 02420, USA.
| | | | - Darrell O Ricke
- Massachusetts Institute of Technology, Lincoln Laboratory, Lexington, MA, 02420, USA.
| |
Collapse
|
86
|
Polimeni JR, Lewis LD. Imaging faster neural dynamics with fast fMRI: A need for updated models of the hemodynamic response. Prog Neurobiol 2021; 207:102174. [PMID: 34525404 PMCID: PMC8688322 DOI: 10.1016/j.pneurobio.2021.102174] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 07/30/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022]
Abstract
Fast fMRI enables the detection of neural dynamics over timescales of hundreds of milliseconds, suggesting it may provide a new avenue for studying subsecond neural processes in the human brain. The magnitudes of these fast fMRI dynamics are far greater than predicted by canonical models of the hemodynamic response. Several studies have established nonlinear properties of the hemodynamic response that have significant implications for fast fMRI. We first review nonlinear properties of the hemodynamic response function that may underlie fast fMRI signals. We then illustrate the breakdown of canonical hemodynamic response models in the context of fast neural dynamics. We will then argue that the canonical hemodynamic response function is not likely to reflect the BOLD response to neuronal activity driven by sparse or naturalistic stimuli or perhaps to spontaneous neuronal fluctuations in the resting state. These properties suggest that fast fMRI is capable of tracking surprisingly fast neuronal dynamics, and we discuss the neuroscientific questions that could be addressed using this approach.
Collapse
Affiliation(s)
- Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA; Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Laura D Lewis
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
87
|
Potjewyd G, Kellett K, Hooper N. 3D hydrogel models of the neurovascular unit to investigate blood-brain barrier dysfunction. Neuronal Signal 2021; 5:NS20210027. [PMID: 34804595 PMCID: PMC8579151 DOI: 10.1042/ns20210027] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/16/2022] Open
Abstract
The neurovascular unit (NVU), consisting of neurons, glial cells, vascular cells (endothelial cells, pericytes and vascular smooth muscle cells (VSMCs)) together with the surrounding extracellular matrix (ECM), is an important interface between the peripheral blood and the brain parenchyma. Disruption of the NVU impacts on blood-brain barrier (BBB) regulation and underlies the development and pathology of multiple neurological disorders, including stroke and Alzheimer's disease (AD). The ability to differentiate induced pluripotent stem cells (iPSCs) into the different cell types of the NVU and incorporate them into physical models provides a reverse engineering approach to generate human NVU models to study BBB function. To recapitulate the in vivo situation such NVU models must also incorporate the ECM to provide a 3D environment with appropriate mechanical and biochemical cues for the cells of the NVU. In this review, we provide an overview of the cells of the NVU and the surrounding ECM, before discussing the characteristics (stiffness, functionality and porosity) required of hydrogels to mimic the ECM when incorporated into in vitro NVU models. We summarise the approaches available to measure BBB functionality and present the techniques in use to develop robust and translatable models of the NVU, including transwell models, hydrogel models, 3D-bioprinting, microfluidic models and organoids. The incorporation of iPSCs either without or with disease-specific genetic mutations into these NVU models provides a platform in which to study normal and disease mechanisms, test BBB permeability to drugs, screen for new therapeutic targets and drugs or to design cell-based therapies.
Collapse
Affiliation(s)
- Geoffrey Potjewyd
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Katherine A.B. Kellett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nigel M. Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, U.K
| |
Collapse
|
88
|
Georgakopoulou T, van der Wijk AE, Bakker ENTP, vanBavel E. Quantitative 3D analysis of tissue damage in a rat model of microembolization. J Biomech 2021; 128:110723. [PMID: 34509910 DOI: 10.1016/j.jbiomech.2021.110723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/16/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
There is a discrepancy between successful recanalization and good clinical outcome after endovascular treatment (EVT) in acute ischemic stroke patients. During removal of a thrombus, a shower of microemboli may release and lodge to the distal circulation. The objective of this study was to determine the extent of damage on brain tissue caused by microemboli. In a rat model of microembolization, a mixture of microsphere (MS) sizes (15, 25 and 50 µm diameter) was injected via the left internal carotid artery. A 3D image of the left hemisphere was reconstructed and a point-pattern spatial analysis was applied based on G- and K-functions to unravel the spatial correlation between MS and the induced hypoxia or infarction. We show a spatial correlation between MS and hypoxia or infarction spreading up to a distance of 1000-1500 µm. These results imply that microemboli, which individually may not always be harmful, can interact and result in local areas of hypoxia or even infarction when lodged in large numbers.
Collapse
Affiliation(s)
- Theodosia Georgakopoulou
- Amsterdam University Medical Centers, University of Amsterdam, Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, The Netherlands
| | - Anne-Eva van der Wijk
- Amsterdam University Medical Centers, University of Amsterdam, Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, The Netherlands
| | - Erik N T P Bakker
- Amsterdam University Medical Centers, University of Amsterdam, Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ed vanBavel
- Amsterdam University Medical Centers, University of Amsterdam, Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, The Netherlands.
| | | |
Collapse
|
89
|
Warner RL, Gast TJ, Sapoznik KA, Carmichael-Martins A, Burns SA. Measuring Temporal and Spatial Variability of Red Blood Cell Velocity in Human Retinal Vessels. Invest Ophthalmol Vis Sci 2021; 62:29. [PMID: 34846516 PMCID: PMC8648047 DOI: 10.1167/iovs.62.14.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The retinal circulation regulates blood flow through various internal and external factors; however, it is unclear how locally these factors act within the retinal microcirculation. We measured the temporal and spatial variability of blood velocity in small retinal vessels using a dual-beam adaptive optics scanning laser ophthalmoscope. Methods In young healthy subjects (n = 3), temporal blood velocity variability was measured in a local vascular region consisting of an arteriole, capillary, and venule repeatedly over 2 days. Data consisted of 10 imaging periods separated into two sessions: (1) five 6-minute image acquisition periods with 30-minute breaks, and (2) five 6-minute image acquisition periods with 10-minute breaks. In another group of young healthy subjects (n = 5), spatial distribution of velocity variability was measured by imaging three capillary segments during three 2-minute conditions: (1) baseline imaging condition (no flicker), (2) full-field flicker, and (3) no flicker condition again. Results Blood velocities were measurable in all subjects with a reliability of about 2%. The coefficient of variation (CV) was used as an estimate of the physiological variability of each vessel. Over 2 days, the average CV in arterioles was 7% (±2%); in capillaries, it was 19% (±6%); and, in venules, it was 8% (±2%). During flicker stimulation, the average capillary CV was 16% during baseline, 15% during flicker stimulation, and 18% after flicker stimulation. Conclusions Capillaries in the human retina exhibit spatial and temporal variations in blood velocity. This inherent variation in blood velocity places limits on studying the vascular regulation of individual capillaries, and the study presented here serves as a foundation for future endeavors.
Collapse
Affiliation(s)
- Raymond L Warner
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Thomas J Gast
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Kaitlyn A Sapoznik
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | | | - Stephen A Burns
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
90
|
Gama Sosa MA, De Gasperi R, Pryor D, Perez Garcia GS, Perez GM, Abutarboush R, Kawoos U, Hogg S, Ache B, Janssen WG, Sowa A, Tetreault T, Cook DG, Tappan SJ, Gandy S, Hof PR, Ahlers ST, Elder GA. Low-level blast exposure induces chronic vascular remodeling, perivascular astrocytic degeneration and vascular-associated neuroinflammation. Acta Neuropathol Commun 2021; 9:167. [PMID: 34654480 PMCID: PMC8518227 DOI: 10.1186/s40478-021-01269-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023] Open
Abstract
Cerebral vascular injury as a consequence of blast-induced traumatic brain injury is primarily the result of blast wave-induced mechanical disruptions within the neurovascular unit. In rodent models of blast-induced traumatic brain injury, chronic vascular degenerative processes are associated with the development of an age-dependent post-traumatic stress disorder-like phenotype. To investigate the evolution of blast-induced chronic vascular degenerative changes, Long-Evans rats were blast-exposed (3 × 74.5 kPa) and their brains analyzed at different times post-exposure by X-ray microcomputed tomography, immunohistochemistry and electron microscopy. On microcomputed tomography scans, regional cerebral vascular attenuation or occlusion was observed as early as 48 h post-blast, and cerebral vascular disorganization was visible at 6 weeks and more accentuated at 13 months post-blast. Progression of the late-onset pathology was characterized by detachment of the endothelial and smooth muscle cellular elements from the neuropil due to degeneration and loss of arteriolar perivascular astrocytes. Development of this pathology was associated with vascular remodeling and neuroinflammation as increased levels of matrix metalloproteinases (MMP-2 and MMP-9), collagen type IV loss, and microglial activation were observed in the affected vasculature. Blast-induced chronic alterations within the neurovascular unit should affect cerebral blood circulation, glymphatic flow and intramural periarterial drainage, all of which may contribute to development of the blast-induced behavioral phenotype. Our results also identify astrocytic degeneration as a potential target for the development of therapies to treat blast-induced brain injury.
Collapse
Affiliation(s)
- Miguel A Gama Sosa
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Georgina S Perez Garcia
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
| | - Gissel M Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, MD, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, MD, USA
| | - Seth Hogg
- Micro Photonics, Inc, 1550 Pond Road, Suite 110, Allentown, PA, 18104, USA
| | - Benjamin Ache
- Micro Photonics, Inc, 1550 Pond Road, Suite 110, Allentown, PA, 18104, USA
| | - William G Janssen
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Allison Sowa
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - David G Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA, 98108, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA
| | - Susan J Tappan
- MBF Bioscience LLC, 185 Allen Brook Lane, Williston, VT, 05495, USA
| | - Sam Gandy
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Patrick R Hof
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Gregory A Elder
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY, 10029, USA
- Mount Sinai Alzheimer's Disease Research Center and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| |
Collapse
|
91
|
Davis CM, Bah TM, Zhang WH, Nelson JW, Golgotiu K, Nie X, Alkayed FN, Young JM, Woltjer RL, Silbert LC, Grafe MR, Alkayed NJ. GPR39 localization in the aging human brain and correlation of expression and polymorphism with vascular cognitive impairment. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12214. [PMID: 34692987 PMCID: PMC8515554 DOI: 10.1002/trc2.12214] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 06/24/2021] [Accepted: 08/11/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The pathogenesis of vascular cognitive impairment (VCI) is not fully understood. GPR39, an orphan G-protein coupled receptor, is implicated in neurological disorders but its role in VCI is unknown. METHODS We performed GPR39 immunohistochemical analysis in post mortem brain samples from mild cognitive impairment (MCI) and control subjects. DNA was analyzed for GPR39 single nucleotide polymorphisms (SNPs), and correlated with white matter hyperintensity (WMH) burden on pre mortem magnetic resonance imaging. RESULTS GPR39 is expressed in aged human dorsolateral prefrontal cortex, localized to microglia and peri-capillary cells resembling pericytes. GPR39-capillary colocalization, and density of GPR39-expressing microglia was increased in aged brains compared to young. SNP distribution was equivalent between groups; however, homozygous SNP carriers were present only in the MCI group, and had higher WMH volume than wild-type or heterozygous SNP carriers. DISCUSSION GPR39 may play a role in aging-related VCI, and may serve as a therapeutic target and biomarker for the risk of developing VCI.
Collapse
Affiliation(s)
- Catherine M. Davis
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Thierno M. Bah
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Wenri H. Zhang
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Jonathan W. Nelson
- Division of Nephrology and Hypertension, Department of MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Kirsti Golgotiu
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Xiao Nie
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Farah N. Alkayed
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Jennifer M. Young
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
- Knight Cardiovascular Institute, Oregon Health & Science UniversityPortlandOregonUSA
| | - Randy L. Woltjer
- Department of PathologyOregon Health & Science UniversityPortlandOregonUSA
| | - Lisa C. Silbert
- Layton Aging and Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - Marjorie R. Grafe
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
- Department of PathologyOregon Health & Science UniversityPortlandOregonUSA
| | - Nabil J. Alkayed
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
- Knight Cardiovascular Institute, Oregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
92
|
Schlecht A, Boneva S, Salie H, Killmer S, Wolf J, Hajdu RI, Auw-Haedrich C, Agostini H, Reinhard T, Schlunck G, Bengsch B, Lange CA. Imaging mass cytometry for high-dimensional tissue profiling in the eye. BMC Ophthalmol 2021; 21:338. [PMID: 34544377 PMCID: PMC8454101 DOI: 10.1186/s12886-021-02099-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/02/2021] [Indexed: 11/10/2022] Open
Abstract
Background Imaging mass cytometry (IMC) combines the principles of flow cytometry and mass spectrometry (MS) with laser scanning spatial resolution and offers unique advantages for the analysis of tissue samples in unprecedented detail. In contrast to conventional immunohistochemistry, which is limited in its application by the number of possible fluorochrome combinations, IMC uses isoptope-coupled antibodies that allow multiplex analysis of up to 40 markers in the same tissue section simultaneously. Methods In this report we use IMC to analyze formalin-fixed, paraffin-embedded conjunctival tissue. We performed a 18-biomarkers IMC analysis of conjunctival tissue to determine and summarize the possibilities, relevance and limitations of IMC for deciphering the biology and pathology of ocular diseases. Results Without modifying the manufacturer’s protocol, we observed positive and plausible staining for 12 of 18 biomarkers. Subsequent bioinformatical single-cell analysis and phenograph clustering identified 24 different cellular clusters with distinct expression profiles with respect to the markers used. Conclusions IMC enables highly multiplexed imaging of ocular samples at subcellular resolution. IMC is an innovative and feasible method, providing new insights into ocular disease pathogenesis that will be valuable for basic research, drug discovery and clinical diagnostics. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-021-02099-8.
Collapse
Affiliation(s)
- Anja Schlecht
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany.,Institute of Anatomy, Wuerzburg University, Wuerzburg, Germany
| | - Stefaniya Boneva
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Henrike Salie
- Faculty of Medicine, Department of Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Saskia Killmer
- Faculty of Medicine, Department of Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Julian Wolf
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Rozina Ida Hajdu
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany.,Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Claudia Auw-Haedrich
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Hansjürgen Agostini
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Thomas Reinhard
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Günther Schlunck
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Bertram Bengsch
- Faculty of Medicine, Department of Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Clemens Ak Lange
- Faculty of Medicine, Eye Center, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany.
| |
Collapse
|
93
|
Miller DR, McClain ES, Dodds JN, Balinski A, May JC, McLean JA, Cliffel DE. Chlorpyrifos Disrupts Acetylcholine Metabolism Across Model Blood-Brain Barrier. Front Bioeng Biotechnol 2021; 9:622175. [PMID: 34513802 PMCID: PMC8431803 DOI: 10.3389/fbioe.2021.622175] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 07/16/2021] [Indexed: 01/25/2023] Open
Abstract
Despite the significant progress in both scientific understanding and regulations, the safety of agricultural pesticides continues to be called into question. The need for complementary analytics to identify dysregulation events associated with chemical exposure and leverage this information to predict biological responses remains. Here, we present a platform that combines a model organ-on-chip neurovascular unit (NVU) with targeted mass spectrometry (MS) and electrochemical analysis to assess the impact of organophosphate (OP) exposure on blood-brain barrier (BBB) function. Using the NVU to simulate exposure, an escalating dose of the organophosphate chlorpyrifos (CPF) was administered. With up to 10 μM, neither CPF nor its metabolites were detected across the BBB (limit of quantitation 0.1 µM). At 30 µM CPF and above, targeted MS detected the main urinary metabolite, trichloropyridinol (TCP), across the BBB (0.025 µM) and no other metabolites. In the vascular chamber where CPF was directly applied, two primary metabolites of CPF, TCP and diethylthiophosphate (DETP), were both detected (0.1–5.7 µM). In a second experiment, a constant dose of 10 µM CPF was administered to the NVU, and though neither CPF nor its metabolites were detected across the BBB after 24 h, electrochemical analysis detected increases in acetylcholine levels on both sides of the BBB (up to 24.8 ± 3.4 µM) and these levels remained high over the course of treatment. In the vascular chamber where CPF was directly applied, only TCP was detected (ranging from 0.06 μM at 2 h to 0.19 μM at 24 h). These results provide chemical evidence of the substantial disruption induced by this widely used commercial pesticide. This work reinforces previously observed OP metabolism and mechanisms of impact, validates the use of the NVU for OP toxicology testing, and provides a model platform for analyzing these organotypic systems.
Collapse
Affiliation(s)
- Dusty R Miller
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Ethan S McClain
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - James N Dodds
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, United States
| | - Andrzej Balinski
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, United States
| | - Jody C May
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, United States
| | - John A McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, United States
| | - David E Cliffel
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
94
|
Eltanahy AM, Koluib YA, Gonzales A. Pericytes: Intrinsic Transportation Engineers of the CNS Microcirculation. Front Physiol 2021; 12:719701. [PMID: 34497540 PMCID: PMC8421025 DOI: 10.3389/fphys.2021.719701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/29/2021] [Indexed: 12/15/2022] Open
Abstract
Pericytes in the brain are candidate regulators of microcirculatory blood flow because they are strategically positioned along the microvasculature, contain contractile proteins, respond rapidly to neuronal activation, and synchronize microvascular dynamics and neurovascular coupling within the capillary network. Analyses of mice with defects in pericyte generation demonstrate that pericytes are necessary for the formation of the blood-brain barrier, development of the glymphatic system, immune homeostasis, and white matter function. The development, identity, specialization, and progeny of different subtypes of pericytes, however, remain unclear. Pericytes perform brain-wide 'transportation engineering' functions in the capillary network, instructing, integrating, and coordinating signals within the cellular communicome in the neurovascular unit to efficiently distribute oxygen and nutrients ('goods and services') throughout the microvasculature ('transportation grid'). In this review, we identify emerging challenges in pericyte biology and shed light on potential pericyte-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ahmed M. Eltanahy
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Yara A. Koluib
- Tanta University Hospitals, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Albert Gonzales
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| |
Collapse
|
95
|
APLN/APLNR Signaling Controls Key Pathological Parameters of Glioblastoma. Cancers (Basel) 2021; 13:cancers13153899. [PMID: 34359800 PMCID: PMC8345670 DOI: 10.3390/cancers13153899] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The neurovascular peptide Apelin and its receptor APLNR are upregulated during glioblastoma pathology. Here we summarize their role in the brain tumor microenvironment composed of neurons, astrocytes, and the vascular and immune systems. Targeting APLN/APLNR signaling promises to unfold multimodal actions in future GBM therapy, acting as an anti-angiogenic and an anti-invasive treatment, and offering the possibility to reduce neurological symptoms and increase overall survival simultaneously. Abstract Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. GBM-expansion depends on a dense vascular network and, coherently, GBMs are highly angiogenic. However, new intratumoral blood vessels are often aberrant with consequences for blood-flow and vascular barrier function. Hence, the delivery of chemotherapeutics into GBM can be compromised. Furthermore, leaky vessels support edema-formation, which can result in severe neurological deficits. The secreted signaling peptide Apelin (APLN) plays an important role in the formation of GBM blood vessels. Both APLN and the Apelin receptor (APLNR) are upregulated in GBM cells and control tumor cell invasiveness. Here we summarize the current evidence on the role of APLN/APLNR signaling during brain tumor pathology. We show that targeting APLN/APLNR can induce anti-angiogenic effects in GBM and simultaneously blunt GBM cell infiltration. In addition, we discuss how manipulation of APLN/APLNR signaling in GBM leads to the normalization of tumor vessels and thereby supports chemotherapy, reduces edema, and improves anti-tumorigenic immune reactions. Hence, therapeutic targeting of APLN/APLNR signaling offers an interesting option to address different pathological hallmarks of GBM.
Collapse
|
96
|
Zhang W, Davis CM, Zeppenfeld DM, Golgotiu K, Wang MX, Haveliwala M, Hong D, Li Y, Wang RK, Iliff JJ, Alkayed NJ. Role of endothelium-pericyte signaling in capillary blood flow response to neuronal activity. J Cereb Blood Flow Metab 2021; 41:1873-1885. [PMID: 33853406 PMCID: PMC8327110 DOI: 10.1177/0271678x211007957] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Local blood flow in the brain is tightly coupled to metabolic demands, a phenomenon termed functional hyperemia. Both capillaries and arterioles contribute to the hyperemic response to neuronal activity via different mechanisms and timescales. The nature and specific signaling involved in the hyperemic response of capillaries versus arterioles, and their temporal relationship are not fully defined. We determined the time-dependent changes in capillary flux and diameter versus arteriolar velocity and flow following whisker stimulation using optical microangiography (OMAG) and two-photon microscopy. We further characterized depth-resolved responses of individual capillaries versus capillary networks. We hypothesized that capillaries respond first to neuronal activation, and that they exhibit a coordinated response mediated via endothelial-derived epoxyeicosatrienoates (EETs) acting on pericytes. To visualize peri-capillary pericytes, we used Tie2-GFP/NG2-DsRed mice, and to determine the role of endothelial-derived EETs, we compared cerebrovascular responses to whisker stimulation between wild-type mice and mice with lower endothelial EETs (Tie2-hsEH). We found that capillaries respond immediately to neuronal activation in an orchestrated network-level manner, a response attenuated in Tie2-hsEH and inhibited by blocking EETs action on pericytes. These results demonstrate that capillaries are first responders during functional hyperemia, and that they exhibit a network-level response mediated via endothelial-derived EETs' action on peri-capillary pericytes.
Collapse
Affiliation(s)
- Wenri Zhang
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Catherine M Davis
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Douglas M Zeppenfeld
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Kirsti Golgotiu
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Marie X Wang
- Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care Center, Seattle, WA, USA.,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Mariya Haveliwala
- Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care Center, Seattle, WA, USA.,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Daniel Hong
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Yuandong Li
- Department of Bioengineering, University of Washington School of Medicine, Seattle, WA, USA
| | - Ruikang K Wang
- Department of Bioengineering, University of Washington School of Medicine, Seattle, WA, USA
| | - Jeffrey J Iliff
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.,Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care Center, Seattle, WA, USA.,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Nabil J Alkayed
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
97
|
Ojo JO, Reed JM, Crynen G, Vallabhaneni P, Evans J, Shackleton B, Eisenbaum M, Ringland C, Edsell A, Mullan M, Crawford F, Bachmeier C. APOE genotype dependent molecular abnormalities in the cerebrovasculature of Alzheimer's disease and age-matched non-demented brains. Mol Brain 2021; 14:110. [PMID: 34238312 PMCID: PMC8268468 DOI: 10.1186/s13041-021-00803-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular dysfunction is a hallmark feature of Alzheimer's disease (AD). One of the greatest risk factors for AD is the apolipoprotein E4 (E4) allele. The APOE4 genotype has been shown to negatively impact vascular amyloid clearance, however, its direct influence on the molecular integrity of the cerebrovasculature compared to other APOE variants (APOE2 and APOE3) has been largely unexplored. To address this, we employed a 10-plex tandem isobaric mass tag approach in combination with an ultra-high pressure liquid chromatography MS/MS (Q-Exactive) method, to interrogate unbiased proteomic changes in cerebrovessels from AD and healthy control brains with different APOE genotypes. We first interrogated changes between healthy control cases to identify underlying genotype specific effects in cerebrovessels. EIF2 signaling, regulation of eIF4 and 70S6K signaling and mTOR signaling were the top significantly altered pathways in E4/E4 compared to E3/E3 cases. Oxidative phosphorylation, EIF2 signaling and mitochondrial dysfunction were the top significant pathways in E2E2 vs E3/E3cases. We also identified AD-dependent changes and their interactions with APOE genotype and found the highest number of significant proteins from this interaction was observed in the E3/E4 (192) and E4/E4 (189) cases. As above, EIF2, mTOR signaling and eIF4 and 70S6K signaling were the top three significantly altered pathways in E4 allele carriers (i.e. E3/E4 and E4/E4 genotypes). Of all the cerebrovascular cell-type specific markers identified in our proteomic analyses, endothelial cell, astrocyte, and smooth muscle cell specific protein markers were significantly altered in E3/E4 cases, while endothelial cells and astrocyte specific protein markers were altered in E4/E4 cases. These proteomic changes provide novel insights into the longstanding link between APOE4 and cerebrovascular dysfunction, implicating a role for impaired autophagy, ER stress, and mitochondrial bioenergetics. These APOE4 dependent changes we identified could provide novel cerebrovascular targets for developing disease modifying strategies to mitigate the effects of APOE4 genotype on AD pathogenesis.
Collapse
Affiliation(s)
- Joseph O Ojo
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA. .,James A. Haley Veterans' Hospital, Tampa, FL, USA. .,The Open University, Milton Keynes, UK.
| | - Jon M Reed
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Gogce Crynen
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA
| | | | - James Evans
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA
| | - Benjamin Shackleton
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Maximillian Eisenbaum
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Charis Ringland
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Anastasia Edsell
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA
| | - Michael Mullan
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK
| | - Fiona Crawford
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,James A. Haley Veterans' Hospital, Tampa, FL, USA.,The Open University, Milton Keynes, UK
| | - Corbin Bachmeier
- Department of Experimental Neuropathology, Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, UK.,Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
98
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
99
|
Terman D, Chen L, Hannawi Y. Mathematical modeling of cerebral capillary blood flow heterogeneity and its effect on brain tissue oxygen levels. J Theor Biol 2021; 527:110817. [PMID: 34157352 DOI: 10.1016/j.jtbi.2021.110817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/06/2021] [Accepted: 06/15/2021] [Indexed: 11/29/2022]
Abstract
Maintaining cerebral blood flow is critical for adequate neuronal function. Previous computational models of brain capillary networks have predicted that heterogeneous cerebral capillary flow patterns result in lower brain tissue partial oxygen pressures PO2). However, these previous models have often considered simple capillary networks in terms of their geometric properties. In this current work, we developed and analyzed computational models of brain capillary networks to determine how perturbations of network properties impact tissue oxygen levels. The models include variabilities in both their geometric (segment lengths and diameters) and three-dimensional, topological structure. Two classes of capillary network models are considered. The first consists of equations for the oxygen partial pressure, PO2, in both a capillary network and the surrounding tissue. In order to gain insight into the behavior of this detailed model, we also consider a reduced model for changes in PO2 in just the capillary network. The main result is that for a general class of networks, random perturbations of either segment diameters or conductances will always, on average, decrease the average tissue oxygen levels. This result is supported through both simulations of the models and mathematical analysis. Our results promise to expand our understanding of cerebral capillary blood flow and its impact on the brain function in health and disease.
Collapse
Affiliation(s)
- David Terman
- Department of Mathematics, The Ohio State University, Columbus, OH 43210 USA.
| | - Liam Chen
- Department of Mathematics, The Ohio State University, Columbus, OH 43210 USA
| | - Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| |
Collapse
|
100
|
Patel B, Yang PH, Kim AH. The effect of thermal therapy on the blood-brain barrier and blood-tumor barrier. Int J Hyperthermia 2021; 37:35-43. [PMID: 32672118 DOI: 10.1080/02656736.2020.1783461] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The blood-brain and blood-tumor barriers represent highly specialized structures responsible for tight regulation of molecular transit into the central nervous system. Under normal circumstances, the relative impermeability of the blood-brain barrier (BBB) protects the brain from circulating toxins and contributes to a brain microenvironment necessary for optimal neuronal function. However, in the context of tumors and other diseases of central nervous system, the BBB and the more recently appreciated blood-tumor barrier (BTB) represent barriers that prevent effective drug delivery. Overcoming both barriers to optimize treatment of central nervous system diseases remains the subject of intense scientific investigation. Although many newer technologies have been developed to overcome these barriers, thermal therapy, which dates back to the 1890 s, has been known to disrupt the BBB since at least the early 1980s. Recently, as a result of several technological advances, laser interstitial thermal therapy (LITT), a method of delivering targeted thermal therapy, has gained widespread use as a surgical technique to ablate brain tumors. In addition, accumulating evidence indicates that laser ablation may also increase local BBB/BTB permeability after treatment. We herein review the structure and function of the BBB and BTB and the impact of thermal injury, including LITT, on barrier function.
Collapse
Affiliation(s)
- Bhuvic Patel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter H Yang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|