1
|
Wellman SM, Forrest AM, Douglas MM, Subbaraman A, Zhang G, Kozai TDY. Dynamic changes in the structure and function of brain mural cells around chronically implanted microelectrodes. Biomaterials 2025; 315:122963. [PMID: 39547137 DOI: 10.1016/j.biomaterials.2024.122963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/25/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Integration of neural interfaces with minimal tissue disruption in the brain is ideal to develop robust tools that can address essential neuroscience questions and combat neurological disorders. However, implantation of intracortical devices provokes severe tissue inflammation within the brain, which requires a high metabolic demand to support a complex series of cellular events mediating tissue degeneration and wound healing. Pericytes, peri-vascular cells involved in blood-brain barrier maintenance, vascular permeability, waste clearance, and angiogenesis, have recently been implicated as potential perpetuators of neurodegeneration in brain injury and disease. While the intimate relationship between pericytes and the cortical microvasculature have been explored in other disease states, their behavior following microelectrode implantation, which is responsible for direct blood vessel disruption and dysfunction, is currently unknown. Using two-photon microscopy we observed dynamic changes in the structure and function of pericytes during implantation of a microelectrode array over a 4-week implantation period. Pericytes respond to electrode insertion through transient increases in intracellular calcium and underlying constriction of capillary vessels. Within days following the initial insertion, we observed an influx of new, proliferating pericytes which contribute to new blood vessel formation. Additionally, we discovered a potentially novel population of reactive immune cells in close proximity to the electrode-tissue interface actively engaging in encapsulation of the microelectrode array. Finally, we determined that intracellular pericyte calcium can be modulated by intracortical microstimulation in an amplitude- and frequency-dependent manner. This study provides a new perspective on the complex biological sequelae occurring at the electrode-tissue interface and will foster new avenues of potential research consideration and lead to development of more advanced therapeutic interventions towards improving the biocompatibility of neural electrode technology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Adam M Forrest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Madeline M Douglas
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ashwat Subbaraman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guangfeng Zhang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Cashion JM, Brown LS, Morris GP, Fortune AJ, Courtney JM, Makowiecki K, Premilovac D, Cullen CL, Young KM, Sutherland BA. Pericyte ablation causes hypoactivity and reactive gliosis in adult mice. Brain Behav Immun 2025; 123:681-696. [PMID: 39406266 DOI: 10.1016/j.bbi.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024] Open
Abstract
Capillary pericytes are important regulators of cerebral blood flow, blood-brain barrier integrity and neuroinflammation, but can become lost or dysfunctional in disease. The consequences of pericyte loss or dysfunction is extremely difficult to discern when it forms one component of a complex disease process. To evaluate this directly, we examined the effect of adult pericyte loss on mouse voluntary movement and motor function, and physiological responses such as hypoxia, blood-brain barrier (BBB) integrity and glial reactivity. Tamoxifen delivery to Pdgfrβ-CreERT2:: Rosa26-DTA transgenic mice was titrated to produce a dose-dependent ablation of pericytes in vivo. 100mg/kg of tamoxifen ablated approximately half of all brain pericytes, while two consecutive daily doses of 300mg/kg tamoxifen ablated >80% of brain pericytes. In the open field test, mice with ∼50% pericyte loss spent more time immobile and travelled half the distance of control mice. Mice with >80% pericyte ablation also slipped more frequently while performing the beam walk task. Our histopathological analyses of the brain revealed that blood vessel density was unchanged, but vessel lumen width was increased. Pericyte-ablated mice also exhibited: mild BBB disruption; increased neuronal hypoxia; astrogliosis and increased IBA1+ immunoreactivity, suggestive of microgliosis and/or macrophage infiltration. Our results highlight the importance of pericytes in the brain, as pericyte loss can directly compromise brain health and induce behavioural alterations in mice.
Collapse
Affiliation(s)
- Jake M Cashion
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Lachlan S Brown
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Gary P Morris
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Alastair J Fortune
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jo-Maree Courtney
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Dino Premilovac
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
3
|
Morton L, Garza AP, Debska-Vielhaber G, Villafuerte LE, Henneicke S, Arndt P, Meuth SG, Schreiber S, Dunay IR. Pericytes and Extracellular Vesicle Interactions in Neurovascular Adaptation to Chronic Arterial Hypertension. J Am Heart Assoc 2024:eJAHA2024038457T. [PMID: 39719419 DOI: 10.1161/jaha.124.038457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Chronic arterial hypertension restructures the vascular architecture of the brain, leading to a series of pathological responses that culminate in cerebral small-vessel disease. Pericytes respond dynamically to vascular challenges; however, how they manifest under the continuous strain of hypertension has not been elucidated. METHODS AND RESULTS In this study, we characterized pericyte behavior alongside hypertensive states in the spontaneously hypertensive stroke-prone rat model, focusing on their phenotypic and metabolic transformation. Flow cytometry was used to characterize pericytes by their expression of platelet-derived growth factor receptor β, neuroglial antigen 2, cluster of differentiation 13-alanyl aminopeptidase, and antigen Kiel 67. Microvessels were isolated for gene expression profiling and in vitro pericyte expansion. Immunofluorescence validated the cell culture model. Plasma-derived extracellular vesicles from hypertensive rodents were applied as a treatment to assess their effects on pericyte function and detailed metabolic assessments on enriched pericytes measured oxidative phosphorylation and glycolysis. Our results reveal a shift in platelet-derived growth factor receptor β+ pericytes toward increased neuroglial antigen 2 and cluster of differentiation 13-alanyl aminopeptidase coexpression, indicative of their critical role in vascular stabilization and inflammatory responses within the hypertensive milieu. Significant alterations were found within key pathways including angiogenesis, blood-brain barrier integrity, hypoxia, and inflammation. Circulating extracellular vesicles from hypertensive rodents distinctly influenced pericyte mitochondrial function, evidencing their dual role as carriers of disease pathology and potential therapeutic agents. Furthermore, a shift toward glycolytic metabolism in hypertensive pericytes was confirmed, coupled with ATP production dysregulation. CONCLUSIONS Our findings demonstrate that cerebral pericytes undergo phenotypic and metabolic reprogramming in response to hypertension, with hypertensive-derived plasma-derived extracellular vesicles impairing their mitochondrial function. Importantly, plasma-derived extracellular vesicles from normotensive controls restore this function, suggesting their potential as both therapeutic agents and precision biomarkers for hypertensive vascular complications. Further investigation into plasma-derived extracellular vesicle cargo is essential to further explore their therapeutic potential in vascular health.
Collapse
Affiliation(s)
- Lorena Morton
- Medical Faculty, Institute of Inflammation and Neurodegeneration Otto-von-Guericke University Magdeburg Magdeburg Germany
| | - Alejandra P Garza
- Medical Faculty, Institute of Inflammation and Neurodegeneration Otto-von-Guericke University Magdeburg Magdeburg Germany
| | | | - Luis E Villafuerte
- Medical Faculty, Institute of Inflammation and Neurodegeneration Otto-von-Guericke University Magdeburg Magdeburg Germany
| | - Solveig Henneicke
- Department of Neurology Otto von Guericke University Magdeburg Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg Magdeburg Germany
| | - Philipp Arndt
- Department of Neurology Otto von Guericke University Magdeburg Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg Magdeburg Germany
| | - Sven G Meuth
- Department of Neurology Heinrich Heine University Düsseldorf Düsseldorf Germany
| | - Stefanie Schreiber
- Department of Neurology Otto von Guericke University Magdeburg Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg Magdeburg Germany
- Center for Behavioral Brain Sciences (CBBS) Magdeburg Germany
- German Center for Mental Health (DZPG) Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C) Halle-Jena-Magdeburg Germany
| | - Ildiko R Dunay
- Medical Faculty, Institute of Inflammation and Neurodegeneration Otto-von-Guericke University Magdeburg Magdeburg Germany
- Center for Behavioral Brain Sciences (CBBS) Magdeburg Germany
- German Center for Mental Health (DZPG) Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C) Halle-Jena-Magdeburg Germany
| |
Collapse
|
4
|
Győri F, Mészáros Á, Krecsmarik M, Molnár K, Balta C, Hermenean A, Farkas AE, Krizbai IA, Wilhelm I. Expression of alpha smooth muscle actin decreases with ageing and increases upon lumen obstruction in mouse brain pericytes. GeroScience 2024:10.1007/s11357-024-01429-0. [PMID: 39592519 DOI: 10.1007/s11357-024-01429-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Cerebral pericytes are mural cells covering brain microvessels, organized as ensheathing, mesh and thin-strand pericytes. These latter two, together called capillary pericytes, have low levels of alpha smooth muscle actin (α-SMA), regulating basal vascular tone and applying a slow influence on cerebral blood flow. Pericytes are subject to alterations in ageing which may be even more pronounced in age-related pathologies, including microinfarcts, which usually affect a large number of vessels in the ageing brain. We modelled this condition by injecting 10 µm-size microspheres into the circulation of mice resulting in the occlusion of capillaries covered by ensheathing and mesh pericytes. We observed that α-SMA and Acta2, the gene encoding it, as well as TGF-β1/Tgfb1, the major regulator of α-SMA, decreased during ageing in cerebral microvessels. In the vicinity of the microspheres stalled in the capillaries, expression of α-SMA increased significantly in both ensheathing and especially in mesh pericytes, both in young (2 to 3 months of age) and old (24 months of age) mice. On the other hand, γ-actin was detected in endothelial cells, but not in pericytes, and decreased in microvessels of microsphere-containing hemispheres. Altogether, our data show that obstruction of cerebral microvessels increases α-SMA expression in pericytes in both age groups, but this does not compensate for the lower expression of the contractile protein in old animals. Increased α-SMA expression may lead to constriction of the obstructed vessels probably aggravating flow heterogeneity in the aged brain.
Collapse
Affiliation(s)
- Fanni Győri
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Ádám Mészáros
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Mónika Krecsmarik
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Cornel Balta
- Aurel Ardelean" Institute of Life Sciences, Vasile Goldiș Western University, Arad, Romania
| | - Anca Hermenean
- Aurel Ardelean" Institute of Life Sciences, Vasile Goldiș Western University, Arad, Romania
| | - Attila E Farkas
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - István A Krizbai
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary.
- Aurel Ardelean" Institute of Life Sciences, Vasile Goldiș Western University, Arad, Romania.
| | - Imola Wilhelm
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary.
- Aurel Ardelean" Institute of Life Sciences, Vasile Goldiș Western University, Arad, Romania.
| |
Collapse
|
5
|
Zhou H, Neudecker V, Perez-Zoghbi JF, Brambrink AM, Yang G. Age-dependent cerebral vasodilation induced by volatile anesthetics is mediated by NG2 + vascular mural cells. Commun Biol 2024; 7:1519. [PMID: 39548262 PMCID: PMC11568297 DOI: 10.1038/s42003-024-07200-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/02/2024] [Indexed: 11/17/2024] Open
Abstract
Anesthesia can influence cerebral blood flow by altering vessel diameter. Using in vivo two-photon imaging, we examined the effects of volatile anesthetics, sevoflurane and isoflurane, on vessel diameter in young and adult mice. Our results show that these anesthetics induce robust dilation of cortical arterioles and arteriole-proximate capillaries in adult mice, with milder effects in juveniles and no dilation in infants. This anesthesia-induced vasodilation correlates with decreased cytosolic Ca2+ levels in NG2+ vascular mural cells. Optogenetic manipulation of these cells bidirectionally regulates vessel diameter, and their ablation abolishes the vasodilatory response to anesthetics. In immature brains, NG2+ mural cells are fewer in number and express lower levels of Kir6.1, a subunit of ATP-sensitive potassium channels. This likely contributes to the age-dependent differences in vasodilation, as Kir6.1 activation promotes, while its inhibition reduces, anesthesia-induced vasodilation. These findings highlight the essential role of NG2+ mural cells in mediating anesthesia-induced cerebral vasodilation.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 100032, USA
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), Shenzhen, Guangdong Province, 518107, China
| | - Viola Neudecker
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 100032, USA
| | - Jose F Perez-Zoghbi
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 100032, USA
| | - Ansgar M Brambrink
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 100032, USA.
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 100032, USA.
| |
Collapse
|
6
|
Cheng C, Deng M, Cheng C, Wu H, Wang Y, Lu M, Yao Z, Li K, Zhang X, Yu B. FOXO1-mTOR pathway in vascular pericyte regulates the formation of type H vessels to control bone metabolism. J Orthop Translat 2024; 49:246-263. [PMID: 39524152 PMCID: PMC11546805 DOI: 10.1016/j.jot.2024.08.010] [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] [Received: 04/09/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 11/16/2024] Open
Abstract
Background As the population aging progresses, age-related osteoporosis has become one of the most common and severe chronic degenerative diseases. Due to insufficient understanding of its complex pathomechanisms, current clinical treatments often suffer from many negative effects. Type H vessels play critical role in bone remodeling owing to their specialized function in coupling angiogenesis and osteogenesis. Increasing evidences have shown a close association between the age-related decline of type H vessels and bone loss. However, the underlying mechanisms whereby the regression of type H vessels with aging remain largely unknown. Methods Col2-Cre ERT /Foxo1 flox/flox mice and FOXO1 inhibitor (AS1842856) treated adult (6 months) and middle aged (10 months) mice were utilized for evaluating the variations in bone volume, bone microarchitecture and type H vessels through micro-CT scanning analysis, histological staining and immunofluorescence staining. In vitro tube-forming and scratch assays were applied to evaluate the angiogenic capacity of human umbilical vein endothelial cells (HUVECs) exposed to AS1842856 or conditioned culture milieu of Human Brain Vascular Pericytes (HBVPs). The expression of pericyte marker proteins, myofibroblast-related proteins and genes in inhibitors-stimulated HBVPs were detected via western blot analysis and Reverse transcription-quantitative PCR (RT-qPCR). Furthermore, perivascular myofibroblastic-like transformation was confirmed in AS1842856-treated animal models through immunofluorescence staining. We also constructed Adipoq-Cre/Foxo1 flox/flox conditional knockout mice and measured their bone mass and type H vessels by micro-CT and immunofluorescence staining. Mechanistic experiments in vitro were conducted via detection of mTOR signalling expression in HBVPs with pharmacological intervention (AS1842856 and rapamycin), genetic knockdown of Foxo1, or FOXO1-overexpression plasmid treatment, verified by RT-qPCR, western blot analysis and cellular immunofluorescence staining. In vivo validation was conducted on Adipoq-Cre/Foxo1 flox/flox mice using immunofluorescence staining. Finally, alterations in osteo-morphology and type H vessels were verified in AS1842856-treated and rapamycin-treated aged mouse models. Results This study identified FOXO1 in pericytes as key components for the formation of type H vessels. We found that FOXO1 expression in pericytes decreases with aging, and pharmacological blocking with AS1842856 promoted type H vessels degeneration and increased bone loss in adult and middle-aged mice, while rapamycin prevented the above pathology in middle-aged mice. We further showed that the loss of FOXO1 in Adipoq + pericytes led to degeneration of type H vessels and bone loss in mice. Mechanistically, the inhibition of FOXO1 by AS1842856 or knockdown of Foxo1 by siRNAs activated mTOR signaling, thereby resulting in the myofibroblastic transformation of pericytes. Furthermore, blocking mTOR signaling by rapamycin rescued the above effects in vitro and in vivo. Conclusion Our findings uncover a hitherto unknown role of FOXO1 in maintaining the phenotype and function of pericytes, thereby promoting formation of type H vessels. This suggests that targeting the FOXO1-mTOR pathway in pericytes could be a potential therapeutic approach to overcome the regression of type H vessels and bone degeneration with aging. The translational potential of this article Our research uncovers a previously unidentified role of FOXO1 in preserving pericyte characteristics and promoting the development of type H vessels. Future translational research targeting the FOXO1-mTOR pathway in pericytes may provide new strategies for the prevention and treatment of age-related osteoporosis in the clinic.
Collapse
Affiliation(s)
- Caiyu Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Mingye Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Chubin Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Hangtian Wu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Yutian Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Mincheng Lu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Zilong Yao
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Kaiqun Li
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| |
Collapse
|
7
|
Suzuki H, Murata J, Unekawa M, Kanno I, Izawa Y, Tomita Y, Tanaka KF, Nakahara J, Masamoto K. Microfluctuations in Capillary Lumens Independent of Pericyte Lining Density in the Anesthetized Mouse Cortex. Microcirculation 2024; 31:e12885. [PMID: 39283679 DOI: 10.1111/micc.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 11/12/2024]
Abstract
OBJECTIVE This study aimed to examine the spatiotemporal coherence of capillary lumen fluctuations in relation to spatial variations in the pericyte lining in the cortex of anesthetized mice. METHODS Two-photon microscopic angiography data (previously published) were reanalyzed, and spatial variations in capillary diameter fluctuations at rest and in capillary lining with vascular mural cells were measured along capillary centerlines. RESULTS Relatively large diameters of the capillaries (5.5 μm) coincided with a dense pericyte lining, while small capillaries (4.3 μm) had a sparse pericyte lining. Temporal variations had a frequency of about 0.1 Hz with an amplitude of 0.5 μm, which were negatively correlated with pericyte lining density. Spatial frequency analysis further revealed a common pattern of spatial variations in capillary diameter and pericyte lining, but temporal variations differed. The temporal variations in capillary lumens were locally distinct from those in neighboring locations, suggesting intrinsic fluctuations independent of the pericyte lining. CONCLUSIONS Capillary lumens in the brain exhibit slow microfluctuations that are independent of pericyte lining. These microfluctuations could affect the distribution of flowing blood cells and may be important for homogenizing their distribution in capillary networks.
Collapse
Affiliation(s)
- Hiroki Suzuki
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Juri Murata
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Tomita Hospital, Nagoya, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuto Masamoto
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
8
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
9
|
Guillot P, Celle S, Barth N, Roche F, Perek N. 'Selected' Exosomes from Sera of Elderly Severe Obstructive Sleep Apnea Patients and Their Impact on Blood-Brain Barrier Function: A Preliminary Report. Int J Mol Sci 2024; 25:11058. [PMID: 39456840 PMCID: PMC11507461 DOI: 10.3390/ijms252011058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) affects a large part of the aging population. It is characterized by chronic intermittent hypoxia and associated with neurocognitive dysfunction. One hypothesis is that the blood-brain barrier (BBB) functions could be altered by exosomes. Exosomes are nanovesicles found in biological fluids. Through the study of exosomes and their content in tau and amyloid beta (Aβ), the aim of this study was to show how exosomes could be used as biomarkers of OSAS and of their cognitive disorders. Two groups of 15 volunteers from the PROOF cohort were selected: severe apnea (AHI > 30) and control (AHI < 5). After exosome isolation from blood serum, we characterized and quantified them (CD81, CD9, CD63) by western blot and ELISAs and put them 5 h in contact with an in vitro BBB model. The apparent permeability of the BBB was measured using sodium-fluorescein and TEER. Cell ELISAs were performed on tight junctions (ZO-1, claudin-5, occludin). The amount of tau and Aβ proteins found in the exosomes was quantified using ELISAs. Compared to controls, OSAS patients had a greater quantity of exosomes, tau, and Aβ proteins in their blood sera, which induced an increase in BBB permeability in the model and was reflected by a loss of tight junction' expression. Elderly patients suffering severe OSAS released more exosomes in serum from the brain compartment than controls. Such exosomes increased BBB permeability. The impact of such alterations on the risk of developing cognitive dysfunction and/or neurodegenerative diseases is questioned.
Collapse
Affiliation(s)
- Pauline Guillot
- Gérontopôle AURA, 42000 Saint-Etienne, France;
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Faculté de Médecine Jacques Lisfranc, Université Jean Monnet, 42000 Saint-Etienne, France
| | - Sebastien Celle
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Physiologie Clinique et de l’Exercice, Centre Visas, CHU Saint Etienne, 42000 Saint-Etienne, France
| | - Nathalie Barth
- Gérontopôle AURA, 42000 Saint-Etienne, France;
- Chaire Santé des Ainés, Ingénierie de la Prévention, Université Jen Monnet, 42000 Saint-Etienne, France
| | - Frederic Roche
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Faculté de Médecine Jacques Lisfranc, Université Jean Monnet, 42000 Saint-Etienne, France
- Physiologie Clinique et de l’Exercice, Centre Visas, CHU Saint Etienne, 42000 Saint-Etienne, France
| | - Nathalie Perek
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Faculté de Médecine Jacques Lisfranc, Université Jean Monnet, 42000 Saint-Etienne, France
| |
Collapse
|
10
|
Esfandi H, Javidan M, Anderson RM, Pashaie R. Depth-Dependent Contributions of Various Vascular Zones to Cerebral Autoregulation and Functional Hyperemia: An In-Silico Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.616950. [PMID: 39416222 PMCID: PMC11482864 DOI: 10.1101/2024.10.07.616950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Autoregulation and neurogliavascular coupling are key mechanisms that modulate myogenic tone (MT) in vessels to regulate cerebral blood flow (CBF) during resting state and periods of increased neural activity, respectively. To determine relative contributions of distinct vascular zones across different cortical depths in CBF regulation, we developed a simplified yet detailed and computationally efficient model of the mouse cerebrovasculature. The model integrates multiple simplifications and generalizations regarding vascular morphology, the hierarchical organization of mural cells, and potentiation/inhibition of MT in vessels. Our analysis showed that autoregulation is the result of the synergy between these factors, but achieving an optimal balance across all cortical depths and throughout the autoregulation range is a complex task. This complexity explains the non-uniformity observed experimentally in capillary blood flow at different cortical depths. In silico simulations of cerebral autoregulation support the idea that the cerebral vasculature does not maintain a plateau of blood flow throughout the autoregulatory range and consists of both flat and sloped phases. We learned that small-diameter vessels with large contractility, such as penetrating arterioles and precapillary arterioles, have major control over intravascular pressure at the entry points of capillaries and play a significant role in CBF regulation. However, temporal alterations in capillary diameter contribute moderately to cerebral autoregulation and minimally to functional hyperemia. In addition, hemodynamic analysis shows that while hemodynamics within capillaries remain relatively stable across all cortical depths throughout the entire autoregulation range, significant variability in hemodynamics can be observed within the first few branch orders of precapillary arterioles or transitional zone vessels. The computationally efficient cerebrovasculature model, proposed in this study, provides a novel framework for analyzing dynamics of the CBF regulation where hemodynamic and vasodynamic interactions are the foundation on which more sophisticated models can be developed.
Collapse
Affiliation(s)
- Hadi Esfandi
- Electrical Engineering and Computer Science Department, Florida Atlantic University, Boca Raton, FL, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mahshad Javidan
- Electrical Engineering and Computer Science Department, Florida Atlantic University, Boca Raton, FL, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Rozalyn M. Anderson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ramin Pashaie
- Electrical Engineering and Computer Science Department, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
11
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
12
|
Bonney SK, Nielson CD, Sosa MJ, Bonnar O, Shih AY. Capillary regression leads to sustained local hypoperfusion by inducing constriction of upstream transitional vessels. Proc Natl Acad Sci U S A 2024; 121:e2321021121. [PMID: 39236241 PMCID: PMC11406265 DOI: 10.1073/pnas.2321021121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
In the brain, a microvascular sensory web coordinates oxygen delivery to regions of neuronal activity. This involves a dense network of capillaries that send conductive signals upstream to feeding arterioles to promote vasodilation and blood flow. Although this process is critical to the metabolic supply of healthy brain tissue, it may also be a point of vulnerability in disease. Deterioration of capillary networks is a feature of many neurological disorders and injuries and how this web is engaged during vascular damage remains unknown. We performed in vivo two-photon microscopy on young adult mural cell reporter mice and induced focal capillary injuries using precise two-photon laser irradiation of single capillaries. We found that ~59% of the injuries resulted in regression of the capillary segment 7 to 14 d following injury, and the remaining repaired to reestablish blood flow within 7 d. Injuries that resulted in capillary regression induced sustained vasoconstriction in the upstream arteriole-capillary transition (ACT) zone at least 21 days postinjury in both awake and anesthetized mice. The degree of vasomotor dynamics was chronically attenuated in the ACT zone consequently reducing blood flow in the ACT zone and in secondary, uninjured downstream capillaries. These findings demonstrate how focal capillary injury and regression can impair the microvascular sensory web and contribute to cerebral hypoperfusion.
Collapse
Affiliation(s)
- Stephanie K. Bonney
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
| | - Cara D. Nielson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| | - Maria J. Sosa
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
| | - Orla Bonnar
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Andy Y. Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
- Department of Pediatrics, University of Washington, Seattle, WA98195
- Department of Bioengineering, University of Washington, Seattle, WA98195
| |
Collapse
|
13
|
Manning D, Rivera EJ, Santana LF. The life cycle of a capillary: Mechanisms of angiogenesis and rarefaction in microvascular physiology and pathologies. Vascul Pharmacol 2024; 156:107393. [PMID: 38857638 DOI: 10.1016/j.vph.2024.107393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Capillaries are the smallest blood vessels (<10 μm in diameter) in the body and their walls are lined by endothelial cells. These microvessels play a crucial role in nutrient and gas exchange between blood and tissues. Capillary endothelial cells also produce vasoactive molecules and initiate the electrical signals that underlie functional hyperemia and neurovascular coupling. Accordingly, capillary function and density are critical for all cell types to match blood flow to cellular activity. This begins with the process of angiogenesis, when new capillary blood vessels emerge from pre-existing vessels, and ends with rarefaction, the loss of these microvascular structures. This review explores the mechanisms behind these processes, emphasizing their roles in various microvascular diseases and their impact on surrounding cells in health and disease. We discuss recent work on the mechanisms controlling endothelial cell proliferation, migration, and tube formation that underlie angiogenesis under physiological and pathological conditions. The mechanisms underlying functional and anatomical rarefaction and the role of pericytes in this process are also discussed. Based on this work, a model is proposed in which the balance of angiogenic and rarefaction signaling pathways in a particular tissue match microvascular density to the metabolic demands of the surrounding cells. This negative feedback loop becomes disrupted during microvascular rarefaction: angiogenic mechanisms are blunted, reactive oxygen species accumulate, capillary function declines and eventually, capillaries disappear. This, we propose, forms the foundation of the reciprocal relationship between vascular density, blood flow, and metabolic needs and functionality of nearby cells.
Collapse
Affiliation(s)
- Declan Manning
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, United States of America.
| | - Ernesto J Rivera
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, United States of America
| | - L Fernando Santana
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, United States of America
| |
Collapse
|
14
|
Edgerton-Fulton M, Abdul Y, Jamil S, Ergul A. Endothelin-1 (ET-1) contributes to senescence and phenotypic changes in brain pericytes in diabetes-mimicking conditions. Clin Sci (Lond) 2024; 138:1009-1022. [PMID: 39106080 DOI: 10.1042/cs20240328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 08/07/2024]
Abstract
Diabetes mediates endothelial dysfunction and increases the risk of Alzheimer's disease and related dementias. Diabetes also dysregulates the ET system. ET-1-mediated constriction of brain microvascular pericytes (BMVPCs) has been shown to contribute to brain hypoperfusion. Cellular senescence, a process that arrests the proliferation of harmful cells and instigates phenotypical changes and proinflammatory responses in endothelial cells that impact their survival and function. Thus, we hypothesized that ET-1 mediates BMVPC senescence and phenotypical changes in diabetes-like conditions. Human BMVPCs were incubated in diabetes-like conditions with or without ET-1 (1 µmol/L) for 3 and 7 days. Hydrogen peroxide (100 µmol/L H2O2) was used as a positive control for senescence and to mimic ischemic conditions. Cells were stained for senescence-associated β-galactosidase or processed for immunoblotting and quantitative real-time PCR analyses. In additional experiments, cells were stimulated with ET-1 in the presence or absence of ETA receptor antagonist BQ-123 (20 μmol/L) or ETB receptor antagonist BQ-788 (20 μmol/L). ET-1 stimulation increased β-galactosidase accumulation which was prevented by BQ-123. ET-1 also increased traditional senescence marker p16 protein and pericyte-specific senescence markers, TGFB1i1, PP1CA, and IGFBP7. Furthermore, ET-1 stimulated contractile protein α-SMA and microglial marker ostepontin in high glucose suggesting a shift toward an ensheathing or microglia-like phenotype. In conclusion, ET-1 triggers senescence, alters ETA and ETB receptors, and causes phenotypical changes in BMVPCs under diabetes-like conditions. These in vitro findings need to be further studied in vivo to establish the role of ETA receptors in the progression of pericyte senescence and phenotypical changes in VCID.
Collapse
Affiliation(s)
- Mia Edgerton-Fulton
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Yasir Abdul
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Sarah Jamil
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Adviye Ergul
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H Johnson VA Health Care System, Medical University of South Carolina, Charleston, SC, U.S.A
| |
Collapse
|
15
|
Liu L, Zhao B, Yu Y, Gao W, Liu W, Chen L, Xia Z, Cao Q. Vascular Aging in Ischemic Stroke. J Am Heart Assoc 2024; 13:e033341. [PMID: 39023057 DOI: 10.1161/jaha.123.033341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cellular senescence, a permanent halt in cell division due to stress, spurs functional and structural changes, contributing to vascular aging characterized by endothelial dysfunction and vascular remodeling. This process raises the risk of ischemic stroke (IS) in older individuals, with its mechanisms still not completely understood despite ongoing research efforts. In this review, we have analyzed the impact of vascular aging on increasing susceptibility and exacerbating the pathology of IS. We have emphasized the detrimental effects of endothelial dysfunction and vascular remodeling influenced by oxidative stress and inflammatory response on vascular aging and IS. Our goal is to aid the understanding of vascular aging and IS pathogenesis, particularly benefiting older adults with high risk of IS.
Collapse
Affiliation(s)
- Lian Liu
- Department of Anesthesiology Renmin Hospital of Wuhan University Wuhan China
| | - Bo Zhao
- Department of Anesthesiology Renmin Hospital of Wuhan University Wuhan China
| | - Yueyang Yu
- Taikang Medical School, School of Basic Medical Sciences Wuhan University Wuhan China
| | - Wenwei Gao
- Department of Critical Care Medicine Renmin Hospital of Wuhan University Wuhan China
| | - Weitu Liu
- Department of Pathology Hubei Provincial Hospital of Traditional Chinese Medicine Wuhan China
| | - Lili Chen
- Department of Anesthesiology Renmin Hospital of Wuhan University Wuhan China
| | - Zhongyuan Xia
- Department of Anesthesiology Renmin Hospital of Wuhan University Wuhan China
| | - Quan Cao
- Department of Nephrology Zhongnan Hospital of Wuhan University Wuhan China
| |
Collapse
|
16
|
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 2024; 15:6398. [PMID: 39080289 PMCID: PMC11289283 DOI: 10.1038/s41467-024-50559-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Aging is frequently associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods and in vivo imaging to determine detailed changes in aged murine cerebrovascular networks. Whole-brain vascular tracing shows an overall ~10% decrease in vascular length and branching density with ~7% increase in vascular radii in aged brains. Light sheet imaging with 3D immunolabeling reveals increased arteriole tortuosity of aged brains. Notably, vasculature and pericyte densities show selective and significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. We find increased blood extravasation, implying compromised blood-brain barrier function in aged brains. Moreover, in vivo imaging in awake mice demonstrates reduced baseline and on-demand blood oxygenation despite relatively intact neurovascular coupling. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Neurosurgery, Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, 94305, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Daniel J Vanselow
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
17
|
Shen K, Shi Y, Wang X, Leung SW. Cellular Components of the Blood-Brain Barrier and Their Involvement in Aging-Associated Cognitive Impairment. Aging Dis 2024:AD.202.0424. [PMID: 39122454 DOI: 10.14336/ad.202.0424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024] Open
Abstract
Human life expectancy has been significantly extended, which poses major challenges to our healthcare and social systems. Aging-associated cognitive impairment is attributed to endothelial dysfunction in the cardiovascular system and neurological dysfunction in the central nervous system. The central nervous system is considered an immune-privileged tissue due to the exquisite protection provided by the blood-brain barrier. The present review provides an overview of the structure and function of blood-brain barrier, extending the cell components of blood-brain barrier from endothelial cells and pericytes to astrocytes, perivascular macrophages and oligodendrocyte progenitor cells. In particular, the pathological changes in the blood-brain barrier in aging, with special focus on the underlying mechanisms and molecular changes, are presented. Furthermore, the potential preventive/therapeutic strategies against aging-associated blood-brain barrier disruption are discussed.
Collapse
Affiliation(s)
- Kaiyuan Shen
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Susan Ws Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Ren SY, Xia Y, Yu B, Lei QJ, Hou PF, Guo S, Wu SL, Liu W, Yang SF, Jiang YB, Chen JF, Shen KF, Zhang CQ, Wang F, Yan M, Ren H, Yang N, Zhang J, Zhang K, Lin S, Li T, Yang QW, Xiao L, Hu ZX, Mei F. Growth hormone promotes myelin repair after chronic hypoxia via triggering pericyte-dependent angiogenesis. Neuron 2024; 112:2177-2196.e6. [PMID: 38653248 DOI: 10.1016/j.neuron.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/26/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
White matter injury (WMI) causes oligodendrocyte precursor cell (OPC) differentiation arrest and functional deficits, with no effective therapies to date. Here, we report increased expression of growth hormone (GH) in the hypoxic neonatal mouse brain, a model of WMI. GH treatment during or post hypoxic exposure rescues hypoxia-induced hypomyelination and promotes functional recovery in adolescent mice. Single-cell sequencing reveals that Ghr mRNA expression is highly enriched in vascular cells. Cell-lineage labeling and tracing identify the GHR-expressing vascular cells as a subpopulation of pericytes. These cells display tip-cell-like morphology with kinetic polarized filopodia revealed by two-photon live imaging and seemingly direct blood vessel branching and bridging. Gain-of-function and loss-of-function experiments indicate that GHR signaling in pericytes is sufficient to modulate angiogenesis in neonatal brains, which enhances OPC differentiation and myelination indirectly. These findings demonstrate that targeting GHR and/or downstream effectors may represent a promising therapeutic strategy for WMI.
Collapse
Affiliation(s)
- Shu-Yu Ren
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Xia
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qi-Jing Lei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng-Fei Hou
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng Guo
- Department of Immunology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Ling Wu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shao-Fan Yang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi-Bin Jiang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kai-Feng Shen
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mi Yan
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China
| | - Hong Ren
- Department of Emergence, 5(th) People's Hospital of Chongqing, Chongqing 400062, China
| | - Nian Yang
- Department of Physiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Zhang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sen Lin
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhang-Xue Hu
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China.
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
19
|
Totoń-Żurańska J, Mikolajczyk TP, Saju B, Guzik TJ. Vascular remodelling in cardiovascular diseases: hypertension, oxidation, and inflammation. Clin Sci (Lond) 2024; 138:817-850. [PMID: 38920058 DOI: 10.1042/cs20220797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
Optimal vascular structure and function are essential for maintaining the physiological functions of the cardiovascular system. Vascular remodelling involves changes in vessel structure, including its size, shape, cellular and molecular composition. These changes result from multiple risk factors and may be compensatory adaptations to sustain blood vessel function. They occur in diverse cardiovascular pathologies, from hypertension to heart failure and atherosclerosis. Dynamic changes in the endothelium, fibroblasts, smooth muscle cells, pericytes or other vascular wall cells underlie remodelling. In addition, immune cells, including macrophages and lymphocytes, may infiltrate vessels and initiate inflammatory signalling. They contribute to a dynamic interplay between cell proliferation, apoptosis, migration, inflammation, and extracellular matrix reorganisation, all critical mechanisms of vascular remodelling. Molecular pathways underlying these processes include growth factors (e.g., vascular endothelial growth factor and platelet-derived growth factor), inflammatory cytokines (e.g., interleukin-1β and tumour necrosis factor-α), reactive oxygen species, and signalling pathways, such as Rho/ROCK, MAPK, and TGF-β/Smad, related to nitric oxide and superoxide biology. MicroRNAs and long noncoding RNAs are crucial epigenetic regulators of gene expression in vascular remodelling. We evaluate these pathways for potential therapeutic targeting from a clinical translational perspective. In summary, vascular remodelling, a coordinated modification of vascular structure and function, is crucial in cardiovascular disease pathology.
Collapse
Affiliation(s)
- Justyna Totoń-Żurańska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz P Mikolajczyk
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Blessy Saju
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| | - Tomasz J Guzik
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
20
|
Cen K, Huang Y, Xie Y, Liu Y. The guardian of intracranial vessels: Why the pericyte? Biomed Pharmacother 2024; 176:116870. [PMID: 38850658 DOI: 10.1016/j.biopha.2024.116870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Intracranial atherosclerotic stenosis (ICAS) is a pathological condition characterized by progressive narrowing or complete blockage of intracranial blood vessels caused by plaque formation. This condition leads to reduced blood flow to the brain, resulting in cerebral ischemia and hypoxia. Ischemic stroke (IS) resulting from ICAS poses a significant global public health challenge, especially among East Asian populations. However, the underlying causes of the notable variations in prevalence among diverse populations, as well as the most effective strategies for preventing and treating the rupture and blockage of intracranial plaques, remain incompletely comprehended. Rupture of plaques, bleeding, and thrombosis serve as precipitating factors in the pathogenesis of luminal obstruction in intracranial arteries. Pericytes play a crucial role in the structure and function of blood vessels and face significant challenges in regulating the Vasa Vasorum (VV)and preventing intraplaque hemorrhage (IPH). This review aims to explore innovative therapeutic strategies that target the pathophysiological mechanisms of vulnerable plaques by modulating pericyte biological function. It also discusses the potential applications of pericytes in central nervous system (CNS) diseases and their prospects as a therapeutic intervention in the field of biological tissue engineering regeneration.
Collapse
Affiliation(s)
- Kuan Cen
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YinFei Huang
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - Yu Xie
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YuMin Liu
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China.
| |
Collapse
|
21
|
Yao Y, Wang Q, Yang J, Yan Y, Wei W. Associations of retinal microvascular alterations with diabetes mellitus: an OCTA-based cross-sectional study. BMC Ophthalmol 2024; 24:245. [PMID: 38858679 PMCID: PMC11165894 DOI: 10.1186/s12886-024-03492-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Diabetes, a health crisis afflicting millions worldwide, is increasing rapidly in prevalence. The microvascular complications triggered by diabetes have emerged as the principal cause of renal disease and blindness. The retinal microvascular network may be sensitive to early systemic vascular structural and functional changes. Therefore, this research endeavored to discern the systemic determinants influencing the retinal microvascular network in patients with and without diabetes. METHODS The Kailuan Eye Study is a cross-sectional study based on the community-based cohort Kailuan Study. Participants underwent optical coherence tomography angiography (OCTA) (Zeiss Cirrus 5000; Carl Zeiss Meditec) and comprehensive systemic examination. Metrics such as perfusion density (PD), vascular density (VD), foveal avascular zone (FAZ) parameters of the superficial capillary plexus (SCP) in the macula were assessed. RESULTS This study included 860 eligible participants (average age = 62.75 ± 6.52 years; 21.9% female), of which 449 were diabetics. People with diabetes had diminished PD and VD in the entire macular and parafoveal regions compared to people without diabetes. Reduced PD in the whole macular region was correlated with higher fasting plasma glucose (FPG, mmol/L) concentration (Beta = -0.19, 95% CI = -0.42 to -0.36, P < 0.001), longer axial length (AL, mm) (Beta = -0.13, 95%CI = -0.48 to -0.25, P = 0.002), and elevated heart rate (Beta = -0.10, 95%CI = -0.14 to -0.19, P = 0.014), after adjusting for younger age (Beta = -0.18, 95%CI = -0.24 to -0.35, P < 0.001), consistent with VD of the whole macular region. A higher FPG level was significantly correlated with lower SCP density of both PD and VD in the macular and parafoveal region (P < 0.05 for all), as well as increased systolic blood pressure and low-density lipoprotein cholesterol concentration (P < 0.01 for all). CONCLUSIONS In this large-sample cross-sectional study, OCTA evaluation revealed that high prevalence of diabetes and elevated FPG levels were correlated with reduced retinal VD and PD. Hypertension and hyperlipidemia are important risk factors for the development of atherosclerotic cardiovascular disease but have no significant effect on retinal microvascular abnormalities.
Collapse
Affiliation(s)
- Yao Yao
- Laboratory of Intraocular Tumor Diagnosis and Treatment, Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijingkey, 100730, China
| | - Qian Wang
- Laboratory of Intraocular Tumor Diagnosis and Treatment, Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijingkey, 100730, China
| | - Jingyan Yang
- Laboratory of Intraocular Tumor Diagnosis and Treatment, Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijingkey, 100730, China
| | - Yanni Yan
- Laboratory of Intraocular Tumor Diagnosis and Treatment, Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijingkey, 100730, China
| | - Wenbin Wei
- Laboratory of Intraocular Tumor Diagnosis and Treatment, Ophthalmology&Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijingkey, 100730, China.
| |
Collapse
|
22
|
Cao Y, Xu W, Liu Q. Alterations of the blood-brain barrier during aging. J Cereb Blood Flow Metab 2024; 44:881-895. [PMID: 38513138 PMCID: PMC11318406 DOI: 10.1177/0271678x241240843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional changes during aging, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. In recent years, advances in microscopy and high-throughput bioinformatics have allowed a more in-depth investigation of the aging mechanisms of BBB. This review summarizes age-related alterations of the BBB structure and function from six perspectives: endothelial cells, astrocytes, pericytes, basement membrane, microglia and perivascular macrophages, and fibroblasts, ranging from the molecular level to the human multi-system level. These basic components are essential for the proper functioning of the BBB. Recent imaging methods of BBB were also reviewed. Elucidation of age-associated BBB changes may offer insights into BBB homeostasis and may provide effective therapeutic strategies to protect it during aging.
Collapse
Affiliation(s)
- Yufan Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weihai Xu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
23
|
Zapata-Acevedo JF, Mantilla-Galindo A, Vargas-Sánchez K, González-Reyes RE. Blood-brain barrier biomarkers. Adv Clin Chem 2024; 121:1-88. [PMID: 38797540 DOI: 10.1016/bs.acc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.
Collapse
Affiliation(s)
- Juan F Zapata-Acevedo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Mantilla-Galindo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
24
|
Miller LR, Bickel MA, Tarantini S, Runion ME, Matacchiera Z, Vance ML, Hibbs C, Vaden H, Nagykaldi D, Martin T, Bullen EC, Pinckard J, Kiss T, Howard EW, Yabluchanskiy A, Conley SM. IGF1R deficiency in vascular smooth muscle cells impairs myogenic autoregulation and cognition in mice. Front Aging Neurosci 2024; 16:1320808. [PMID: 38425784 PMCID: PMC10902040 DOI: 10.3389/fnagi.2024.1320808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Cerebrovascular pathologies contribute to cognitive decline during aging, leading to vascular cognitive impairment and dementia (VCID). Levels of circulating insulin-like growth factor 1 (IGF-1), a vasoprotective hormone, decrease during aging. Decreased circulating IGF-1 in animal models leads to the development of VCID-like symptoms, but the cellular mechanisms underlying IGF-1-deficiency associated pathologies in the aged cerebrovasculature remain poorly understood. Here, we test the hypothesis that vascular smooth muscle cells (VSMCs) play an integral part in mediating the vasoprotective effects of IGF-1. Methods We used a hypertension-based model of cerebrovascular dysfunction in mice with VSMC-specific IGF-1 receptor (Igf1r) deficiency and evaluated the development of cerebrovascular pathologies and cognitive dysfunction. Results VSMC-specific Igf1r deficiency led to impaired cerebral myogenic autoregulation, independent of blood pressure changes, which was also associated with impaired spatial learning and memory function as measured by radial arm water maze and impaired motor learning measured by rotarod. In contrast, VSMC-specific IGF-1 receptor knockdown did not lead to cerebral microvascular rarefaction. Discussion These studies suggest that VSMCs are key targets for IGF-1 in the context of cerebrovascular health, playing a role in vessel stability alongside other cells in the neurovascular unit, and that VSMC dysfunction in aging likely contributes to VCID.
Collapse
Affiliation(s)
- Lauren R. Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Megan E. Runion
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Zoe Matacchiera
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michaela L. Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Clara Hibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hannah Vaden
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Domonkos Nagykaldi
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Teryn Martin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Elizabeth C. Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jessica Pinckard
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Tamas Kiss
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
| | - Eric W. Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
25
|
Stamenkovic S, Schmid F, Weitermann N, Takasaki K, Bonney SK, Sosa MJ, Li Y, Bennett HC, Kim Y, Waters J, Shih AY. Impaired drainage through capillary-venous networks contributes to age-related white matter loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.11.579849. [PMID: 38405879 PMCID: PMC10888936 DOI: 10.1101/2024.02.11.579849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The gradual loss of cerebral white matter contributes to cognitive decline during aging. However, microvascular networks that support the metabolic demands of white matter remain poorly defined. We used in vivo deep multi-photon imaging to characterize microvascular networks that perfuse cortical layer 6 and corpus callosum, a highly studied region of white matter in the mouse brain. We show that these deep tissues are exclusively drained by sparse and wide-reaching venules, termed principal cortical venules, which mirror vascular architecture at the human cortical-U fiber interface. During aging, capillary networks draining into deep branches of principal cortical venules are selectively constricted, reduced in density, and diminished in pericyte numbers. This causes hypo-perfusion in deep tissues, and correlates with gliosis and demyelination, whereas superficial tissues become relatively hyper-perfused. Thus, age-related impairment of capillary-venular drainage is a key vascular deficit that contributes to the unique vulnerability of cerebral white matter during brain aging.
Collapse
|
26
|
Ma Y, Sun W, Bai J, Gao F, Ma H, Liu H, Hu J, Xu C, Zhang X, Liu Z, Yuan T, Sun C, Huang Y, Wang R. Targeting blood brain barrier-Remote ischemic conditioning alleviates cognitive impairment in female APP/PS1 rats. CNS Neurosci Ther 2024; 30:e14613. [PMID: 38379185 PMCID: PMC10879645 DOI: 10.1111/cns.14613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/16/2023] [Accepted: 11/26/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a significant global health concern, and it is crucial that we find effective methods to prevent or slow down AD progression. Recent studies have highlighted the essential role of blood vessels in clearing Aβ, a protein that contributes to AD. Scientists are exploring blood biomarkers as a potential tool for future AD diagnosis. One promising method that may help prevent AD is remote ischemic conditioning (RIC). RIC involves using sub-lethal ischemic-reperfusion cycles on limbs. However, a comprehensive understanding of how RIC can prevent AD and its long-term effectiveness is still lacking. Further research is essential to fully comprehend the potential benefits of RIC in preventing AD. METHODS Female wild-type (WT) and APP/PS1 transgenic rats, aged 12 months, underwent ovariectomy and were subsequently assigned to WT, APP/PS1, and APP/PS1 + RIC groups. RIC was conducted five times a week for 4 weeks. The rats' depressive and cognitive behaviors were evaluated using force swimming, open-field tests, novel objective recognition, elevated plus maze, and Barnes maze tests. Evaluation of the neurovascular unit (NVU), synapses, vasculature, astrocytes, and microglia was conducted using immunofluorescence staining (IF), Western blot (WB), and transmission electron microscopy (TEM). Additionally, the cerebro-vasculature was examined using micro-CT, and cerebral blood flow (CBF) was measured using Speckle Doppler. Blood-brain barrier (BBB) permeability was determined by measuring the Evans blue leakage. Finally, Aβ levels in the rat frontal cortex were measured using WB, ELISA, or IF staining. RESULTS RIC enhanced memory-related protein expression and rescued depressive-like behavior and cognitive decline in APP/PS1 transgenic rats. Additionally, the intervention protected NVU in the rat frontal cortex, as evidenced by (1) increased expression of TJ (tight junction) proteins, pericyte marker PDGFRβ, and glucose transporter 1 (GLUT1), as well as decreased VCAM1; (2) mitigation of ultrastructure impairment in neuron, cerebral vascular, and astrocyte; (3) upregulation of A2 astrocyte phenotype markers and downregulation of A1 phenotype markers, indicating a shift toward a healthier phenotype. Correspondingly, RIC intervention alleviated neuroinflammation, as evidenced by the decreased Iba1 level, a microglia marker. Meanwhile, RIC intervention elevated CBF in frontal cortex of the rats. Notably, RIC intervention effectively suppressed Aβ toxicity, as demonstrated by the enhancement of α-secretase and attenuation of β-secretase (BACE1) and γ- secretase and Aβ1-42 and Aβ1-40 levels as well. CONCLUSION Chronic RIC intervention exerts vascular and neuroprotective roles, suggesting that RIC could be a promising therapeutic strategy targeting the BBB and NVU during AD development.
Collapse
Affiliation(s)
- Yuxuan Ma
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Wuxiang Sun
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Jing Bai
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Fujia Gao
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Haoran Ma
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Huiyu Liu
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Jiewei Hu
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Chao Xu
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Xin Zhang
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Zixuan Liu
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Tao Yuan
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
| | - Chenxu Sun
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Yuanyuan Huang
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| | - Ruimin Wang
- International Science & Technology Cooperation Base of GeriatricSchool of Public Health of North China University of Science and TechnologyTangshanHebeiChina
- School of Basic Medical ScienceNorth China University of Science and TechnologyTangshanHebeiChina
| |
Collapse
|
27
|
van Splunder H, Villacampa P, Martínez-Romero A, Graupera M. Pericytes in the disease spotlight. Trends Cell Biol 2024; 34:58-71. [PMID: 37474376 PMCID: PMC10777571 DOI: 10.1016/j.tcb.2023.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 07/22/2023]
Abstract
Pericytes are known as the mural cells in small-caliber vessels that interact closely with the endothelium. Pericytes play a key role in vasculature formation and homeostasis, and when dysfunctional contribute to vasculature-related diseases such as diabetic retinopathy and neurodegenerative conditions. In addition, significant extravascular roles of pathological pericytes are being discovered with relevant implications for cancer and fibrosis. Pericyte research is challenged by the lack of consistent molecular markers and clear discrimination criteria versus other (mural) cells. However, advances in single-cell approaches are uncovering and clarifying mural cell identities, biological functions, and ontogeny across organs. We discuss the latest developments in pericyte pathobiology to inform future research directions and potential outcomes.
Collapse
Affiliation(s)
- Hielke van Splunder
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Pilar Villacampa
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute (IDIBELL), Carrer de la Feixa Llarga s/n, 08907 l'Hospitalet de Llobregat, Barcelona, Spain
| | - Anabel Martínez-Romero
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain; Institución Catalana de Investigación y Estudios Avanzados (ICREA), Passeig de Lluís Companys 23, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Avenida de Monforte de Lemos 5, 28029 Madrid, Spain.
| |
Collapse
|
28
|
Naranjo O, Torices S, Clifford PR, Rodriguez T, Osborne OM, Tiburcio D, Fattakhov N, Park M, Stevenson M, Toborek M. AKT signaling modulates latent viral reservoir viability in HIV-1-infected blood-brain barrier pericytes. J Biol Chem 2024; 300:105526. [PMID: 38043797 PMCID: PMC10777012 DOI: 10.1016/j.jbc.2023.105526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023] Open
Abstract
Despite antiretroviral therapy (ART), chronic forms of HIV-associated neurocognitive disorders (HAND) affect an estimated 50% of individuals living with HIV, greatly impacting their quality of life. The prevailing theory of HAND progression posits that chronic inflammation arising from the activation of latent viral reservoirs leads to progressive damage in the central nervous system (CNS). Recent evidence indicates that blood-brain barrier (BBB) pericytes are capable of active HIV-1 infection; however, their latent infection has not been defined. Given their location and function, BBB pericytes are poised to be a key viral reservoir in the development of HAND. We present the first transcriptional analysis of uninfected, active, and latent human BBB pericytes, revealing distinct transcriptional phenotypes. In addition, we demonstrate that latent infection of BBB pericytes relies on AKT signaling for reservoir survival. These findings provide insight into the state of reservoir maintenance in the CNS during HIV-1 infection and provide novel targets for reservoir clearance.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA.
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Paul R Clifford
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Thaidy Rodriguez
- Department of Urology, University of California San Francisco, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Destiny Tiburcio
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Minseon Park
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Mario Stevenson
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
29
|
Ronaldson PT, Williams EI, Betterton RD, Stanton JA, Nilles KL, Davis TP. CNS Drug Delivery in Stroke: Improving Therapeutic Translation From the Bench to the Bedside. Stroke 2024; 55:190-202. [PMID: 38134249 PMCID: PMC10752297 DOI: 10.1161/strokeaha.123.043764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Drug development for ischemic stroke is challenging as evidenced by the paucity of therapeutics that have advanced beyond a phase III trial. There are many reasons for this lack of clinical translation including factors related to the experimental design of preclinical studies. Often overlooked in therapeutic development for ischemic stroke is the requirement of effective drug delivery to the brain, which is critical for neuroprotective efficacy of several small and large molecule drugs. Advancing central nervous system drug delivery technologies implies a need for detailed comprehension of the blood-brain barrier (BBB) and neurovascular unit. Such knowledge will permit the innate biology of the BBB/neurovascular unit to be leveraged for improved bench-to-bedside translation of novel stroke therapeutics. In this review, we will highlight key aspects of BBB/neurovascular unit pathophysiology and describe state-of-the-art approaches for optimization of central nervous system drug delivery (ie, passive diffusion, mechanical opening of the BBB, liposomes/nanoparticles, transcytosis, intranasal drug administration). Additionally, we will discuss how endogenous BBB transporters represent the next frontier of drug delivery strategies for stroke. Overall, this review will provide cutting edge perspective on how central nervous system drug delivery must be considered for the advancement of new stroke drugs toward human trials.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Erica I Williams
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Robert D Betterton
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Joshua A Stanton
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Kelsy L Nilles
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| |
Collapse
|
30
|
Ronaldson PT, Davis TP. Blood-brain barrier transporters: a translational consideration for CNS delivery of neurotherapeutics. Expert Opin Drug Deliv 2024; 21:71-89. [PMID: 38217410 PMCID: PMC10842757 DOI: 10.1080/17425247.2024.2306138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Successful neuropharmacology requires optimization of CNS drug delivery and, by extension, free drug concentrations at brain molecular targets. Detailed assessment of blood-brain barrier (BBB) physiological characteristics is necessary to achieve this goal. The 'next frontier' in CNS drug delivery is targeting BBB uptake transporters, an approach that requires evaluation of brain endothelial cell transport processes so that effective drug accumulation and improved therapeutic efficacy can occur. AREAS COVERED BBB permeability of drugs is governed by tight junction protein complexes (i.e., physical barrier) and transporters/enzymes (i.e., biochemical barrier). For most therapeutics, a component of blood-to-brain transport involves passive transcellular diffusion. Small molecule drugs that do not possess acceptable physicochemical characteristics for passive permeability may utilize putative membrane transporters for CNS uptake. While both uptake and efflux transport mechanisms are expressed at the brain microvascular endothelium, uptake transporters can be targeted for optimization of brain drug delivery and improved treatment of neurological disease states. EXPERT OPINION Uptake transporters represent a unique opportunity to optimize brain drug delivery by leveraging the endogenous biology of the BBB. A rigorous understanding of these transporters is required to improve translation from the bench to clinical trials and stimulate the development of new treatment paradigms for neurological diseases.
Collapse
Affiliation(s)
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona College of Medicine
| |
Collapse
|
31
|
Fang X, Fan F, Border JJ, Roman RJ. Cerebrovascular Dysfunction in Alzheimer's Disease and Transgenic Rodent Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2024; 5:42-64. [PMID: 38434588 PMCID: PMC10906803 DOI: 10.33696/neurol.5.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Alzheimer's Disease (AD) and Alzheimer's Disease-Related Dementia (ADRD) are the primary causes of dementia that has a devastating effect on the quality of life and is a tremendous economic burden on the healthcare system. The accumulation of extracellular beta-amyloid (Aβ) plaques and intracellular hyperphosphorylated tau-containing neurofibrillary tangles (NFTs) in the brain are the hallmarks of AD. They are also thought to be the underlying cause of inflammation, neurodegeneration, brain atrophy, and cognitive impairments that accompany AD. The discovery of APP, PS1, and PS2 mutations that increase Aβ production in families with early onset familial AD led to the development of numerous transgenic rodent models of AD. These models have provided new insight into the role of Aβ in AD; however, they do not fully replicate AD pathology in patients. Familial AD patients with mutations that elevate the production of Aβ represent only a small fraction of dementia patients. In contrast, those with late-onset sporadic AD constitute the majority of cases. This observation, along with the failure of previous clinical trials targeting Aβ or Tau and the modest success of recent trials using Aβ monoclonal antibodies, has led to a reappraisal of the view that Aβ accumulation is the sole factor in the pathogenesis of AD. More recent studies have established that cerebral vascular dysfunction is one of the earliest changes seen in AD, and 67% of the candidate genes linked to AD are expressed in the cerebral vasculature. Thus, there is an increasing appreciation of the vascular contribution to AD, and the National Institute on Aging (NIA) and the Alzheimer's Disease Foundation recently prioritized it as a focused research area. This review summarizes the strengths and limitations of the most commonly used transgenic AD animal models and current views about the contribution of Aβ accumulation versus cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Physiology, Augusta University, Augusta, GA 30912, USA
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
32
|
You TY, Dong Q, Cui M. Emerging Links between Cerebral Blood Flow Regulation and Cognitive Decline: A Role for Brain Microvascular Pericytes. Aging Dis 2023:AD.2022.1204. [PMID: 37163446 PMCID: PMC10389833 DOI: 10.14336/ad.2022.1204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/04/2022] [Indexed: 05/12/2023] Open
Abstract
Cognitive impairment associated with vascular etiology has been of considerable interest in the development of dementia. Recent studies have started to uncover cerebral blood flow deficits in initiating cognitive deterioration. Brain microvascular pericytes, the only type of contractile cells in capillaries, are involved in the precise modulation of vascular hemodynamics due to their ability to regulate resistance in the capillaries. They exhibit potential in maintaining the capillary network geometry and basal vascular tone. In addition, pericytes can facilitate better blood flow supply in response to neurovascular coupling. Their dysfunction is thought to disturb cerebral blood flow causing metabolic imbalances or structural injuries, leading to consequent cognitive decline. In this review, we summarize the characteristics of microvascular pericytes in brain blood flow regulation and outline the framework of a two-hit hypothesis in cognitive decline, where we emphasize how pericytes serve as targets of cerebral blood flow dysregulation that occurs with neurological challenges, ranging from genetic factors, aging, and pathological proteins to ischemic stress.
Collapse
Affiliation(s)
- Tong-Yao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Brown LS, King NE, Courtney JM, Gasperini RJ, Foa L, Howells DW, Sutherland BA. Brain pericytes in culture display diverse morphological and functional phenotypes. Cell Biol Toxicol 2023; 39:2999-3014. [PMID: 37322257 PMCID: PMC10693527 DOI: 10.1007/s10565-023-09814-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/23/2023] [Indexed: 06/17/2023]
Abstract
Pericytes play several important functions in the neurovascular unit including contractile control of capillaries, maintenance of the BBB, regulation of angiogenesis, and neuroinflammation. There exists a continuum of pericyte subtypes along the vascular tree which exhibit both morphological and transcriptomic differences. While different functions have been associated with the pericyte subtypes in vivo, numerous recent publications have used a primary human brain vascular pericytes (HBVP) cell line where this pericyte heterogeneity has not been considered. Here, we used primary HBVP cultures, high-definition imaging, cell motility tracking, and immunocytochemistry to characterise morphology, protein expression, and contractile behaviour to determine whether heterogeneity of pericytes also exists in cultures. We identified five distinct morphological subtypes that were defined using both qualitative criteria and quantitative shape analysis. The proportion of each subtype present within the culture changed as passage number increased, but pericytes did not change morphological subtype over short time periods. The rate and extent of cellular and membrane motility differed across the subtypes. Immunocytochemistry revealed differential expression of alpha-smooth muscle actin (αSMA) across subtypes. αSMA is essential for cell contractility, and consequently, only subtypes with high αSMA expression contracted in response to physiological vasoconstrictors endothelin-1 (ET1) and noradrenaline (NA). We conclude that there are distinct morphological subtypes in HBVP culture, which display different behaviours. This has significance for the use of HBVP when modelling pericyte physiology in vitro where relevance to in vivo pericyte subtypes along the vascular tree must be considered.
Collapse
Affiliation(s)
- Lachlan S Brown
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia
| | - Natalie E King
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia
| | - Jo-Maree Courtney
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia
| | - Robert J Gasperini
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia
| | - Lisa Foa
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia
- School of Psychological Sciences, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - David W Howells
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Level 4 Medical Sciences Precinct, 17 Liverpool St, Hobart, TAS, 7000, Australia.
| |
Collapse
|
34
|
Liang J, Zhao J, Chen Y, Li B, Li Y, Lu F, Dong Z. New Insights and Advanced Strategies for In Vitro Construction of Vascularized Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:692-709. [PMID: 37409413 DOI: 10.1089/ten.teb.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Inadequate vascularization is a significant barrier to clinical application of large-volume tissue engineered grafts. In contrast to in vivo vascularization, in vitro prevascularization shortens the time required for host vessels to grow into the graft core and minimizes necrosis in the core region of the graft. However, the challenge of prevascularization is to construct hierarchical perfusable vascular networks, increase graft volume, and form a vascular tip that can anastomose with host vessels. Understanding advances in in vitro prevascularization techniques and new insights into angiogenesis could overcome these obstacles. In the present review, we discuss new perspectives on angiogenesis, the differences between in vivo and in vitro tissue vascularization, the four elements of prevascularized constructs, recent advances in perfusion-based in vitro prevascularized tissue fabrication, and prospects for large-volume prevascularized tissue engineering.
Collapse
Affiliation(s)
- Jiancong Liang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jing Zhao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yunzi Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Bin Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ziqing Dong
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
35
|
Bonney SK, Nielson CD, Sosa MJ, Shih AY. Capillary regression leads to sustained local hypoperfusion by inducing constriction of upstream transitional vessels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.28.564529. [PMID: 37961686 PMCID: PMC10635020 DOI: 10.1101/2023.10.28.564529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In the brain, a microvascular sensory web coordinates oxygen delivery to regions of neuronal activity. This involves a dense network of capillaries that send conductive signals upstream to feeding arterioles to promote vasodilation and blood flow. Although this process is critical to the metabolic supply of healthy brain tissue, it may also be a point of vulnerability in disease. Deterioration of capillary networks is a hallmark of many neurological disorders and how this web is engaged during vascular damage remains unknown. We performed in vivo two-photon microscopy on young adult mural cell reporter mice and induced focal capillary injuries using precise two-photon laser irradiation of single capillaries. We found that ∼63% of the injuries resulted in regression of the capillary segment 7-14 days following injury, and the remaining repaired to re-establish blood flow within 7 days. Injuries that resulted in capillary regression induced sustained vasoconstriction in the upstream arteriole-capillary transition (ACT) zone at least 21 days post-injury in both awake and anesthetized mice. This abnormal vasoconstriction involved attenuation of vasomotor dynamics and uncoupling from mural cell calcium signaling following capillary regression. Consequently, blood flow was reduced in the ACT zone and in secondary, uninjured downstream capillaries. These findings demonstrate how capillary injury and regression, as often seen in age-related neurological disease, can impair the microvascular sensory web and contribute to cerebral hypoperfusion. SIGNIFICANCE Deterioration of the capillary network is a characteristic of many neurological diseases and can exacerbate neuronal dysfunction and degeneration due to poor blood perfusion. Here we show that focal capillary injuries can induce vessel regression and elicit sustained vasoconstriction in upstream transitional vessels that branch from cortical penetrating arterioles. This reduces blood flow to broader, uninjured regions of the same microvascular network. These findings suggest that widespread and cumulative damage to brain capillaries in neurological disease may broadly affect blood supply and contribute to hypoperfusion through their remote actions.
Collapse
|
36
|
Shi X. Research advances in cochlear pericytes and hearing loss. Hear Res 2023; 438:108877. [PMID: 37651921 PMCID: PMC10538405 DOI: 10.1016/j.heares.2023.108877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/03/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Pericytes are specialized mural cells surrounding endothelial cells in microvascular beds. They play a role in vascular development, blood flow regulation, maintenance of blood-tissue barrier integrity, and control of angiogenesis, tissue fibrosis, and wound healing. In recent decades, understanding of the critical role played by pericytes in retina, brain, lung, and kidney has seen significant progress. The cochlea contains a large population of pericytes. However, the role of cochlear pericytes in auditory pathophysiology is, by contrast, largely unknown. The present review discusses recent progress in identifying cochlear pericytes, mapping their distribution, and defining their role in regulating blood flow, controlling the blood-labyrinth barrier (BLB) and angiogenesis, and involvement in different types of hearing loss.
Collapse
Affiliation(s)
- Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center (NRC04), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| |
Collapse
|
37
|
Fesharaki NJ, Taylor A, Mosby K, Kim JH, Ress D. Global effects of aging on the hemodynamic response function in the human brain. RESEARCH SQUARE 2023:rs.3.rs-3299293. [PMID: 37720046 PMCID: PMC10503846 DOI: 10.21203/rs.3.rs-3299293/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
In functional magnetic resonance imaging, the hemodynamic response function (HRF) is a transient, stereotypical response to local changes in cerebral hemodynamics and oxygen metabolism due to briefly (< 4 s) evoked neural activity. Accordingly, the HRF is often used as an impulse response with the assumption of linearity in data analysis. In cognitive aging studies, it has been very common to interpret differences in brain activation as age-related changes in neural activity. Contrary to this assumption, however, evidence has accrued that normal aging may also significantly affect the vasculature, thereby affecting cerebral hemodynamics and metabolism, confounding interpretation of fMRI aging studies. In this study, use was made of a multisensory stimulus to evoke the HRF in ~ 87% of cerebral cortex in cognitively intact adults with ages ranging from 22-75 years. The stimulus evokes both positive and negative HRFs, which were characterized using model-free parameters in native-space coordinates. Results showed significant age trends in HRF parameter distributions in terms of both amplitudes (e.g., peak amplitude and CNR) and temporal dynamics (e.g., full-width-at-half-maximum). This work sets the stage for using HRF methods as a biomarker for age-related pathology.
Collapse
|
38
|
Zhou R, Li J, Wang R, Chen Z, Zhou F. The neurovascular unit in healthy and injured spinal cord. J Cereb Blood Flow Metab 2023; 43:1437-1455. [PMID: 37190756 PMCID: PMC10414016 DOI: 10.1177/0271678x231172008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/09/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023]
Abstract
The neurovascular unit (NVU) reflects the close temporal and spatial link between neurons and blood vessels. However, the understanding of the NVU in the spinal cord is far from clear and largely based on generalized knowledge obtained from the brain. Herein, we review the present knowledge of the NVU and highlight candidate approaches to investigate the NVU, particularly focusing on the spinal cord. Several unique features maintain the highly regulated microenvironment in the NVU. Autoregulation and neurovascular coupling ensure regional blood flow meets the metabolic demand according to the blood supply or local neural activation. The blood-central nervous system barrier partitions the circulating blood from neural parenchyma and facilitates the selective exchange of substances. Furthermore, we discuss spinal cord injury (SCI) as a common injury from the perspective of NVU dysfunction. Hopefully, this review will help expand the understanding of the NVU in the spinal cord and inspire new insights into SCI.
Collapse
Affiliation(s)
- Rubing Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Junzhao Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
39
|
Khaing ZZ, Chandrasekaran A, Katta A, Reed MJ. The Brain and Spinal Microvasculature in Normal Aging. J Gerontol A Biol Sci Med Sci 2023; 78:1309-1319. [PMID: 37093786 PMCID: PMC10395569 DOI: 10.1093/gerona/glad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Indexed: 04/25/2023] Open
Abstract
Changes in the brain and spinal cord microvasculature during normal aging contribute to the "sensitive" nature of aged central nervous system tissue to ischemic insults. In this review, we will examine alterations in the central nervous system microvasculature during normal aging, which we define as aging without a dominant pathology such as neurodegenerative processes, vascular injury or disease, or trauma. We will also discuss newer technologies to improve the study of central nervous system microvascular structure and function. Microvasculature within the brain and spinal cord will be discussed separately as anatomy and physiology differ between these compartments. Lastly, we will identify critical areas for future studies as well as key unanswered questions.
Collapse
Affiliation(s)
- Zin Z Khaing
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | | | - Anjali Katta
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | - May J Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
40
|
Murata J, Unekawa M, Kudo Y, Kotani M, Kanno I, Izawa Y, Tomita Y, Tanaka KF, Nakahara J, Masamoto K. Acceleration of the Development of Microcirculation Embolism in the Brain due to Capillary Narrowing. Stroke 2023; 54:2135-2144. [PMID: 37309687 DOI: 10.1161/strokeaha.122.042416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/05/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Cerebral microvascular obstruction is critically involved in recurrent stroke and decreased cerebral blood flow with age. The obstruction must occur in the capillary with a greater resistance to perfusion pressure through the microvascular networks. However, little is known about the relationship between capillary size and embolism formation. This study aimed to determine whether the capillary lumen space contributes to the development of microcirculation embolism. METHODS To spatiotemporally manipulate capillary diameters in vivo, transgenic mice expressing the light-gated cation channel protein ChR2 (channelrhodopsin-2) in mural cells were used. The spatiotemporal changes in the regional cerebral blood flow in response to the photoactivation of ChR2 mural cells were first characterized using laser speckle flowgraphy. Capillary responses to optimized photostimulation were then examined in vivo using 2-photon microscopy. Finally, microcirculation embolism due to intravenously injected fluorescent microbeads was compared under conditions with or without photoactivation of ChR2 mural cells. RESULTS Following transcranial photostimulation, the stimulation intensity-dependent decrease in cerebral blood flow centered at the irradiation was observed (14%-49% decreases relative to the baseline). The cerebrovascular response to photostimulation showed significant constriction of the cerebral arteries and capillaries but not of the veins. As a result of vasoconstriction, a temporal stall of red blood cell flow occurred in the capillaries of the venous sides. The 2-photon excitation of a single ChR2 pericyte demonstrated the partial shrinkage of capillaries (7% relative to the baseline) around the stimulated cell. With the intravenous injection of microbeads, the occurrence of microcirculation embolism was significantly enhanced (11% increases compared to the control) with photostimulation. CONCLUSIONS Capillary narrowing increases the risk of developing microcirculation embolism in the venous sides of the cerebral capillaries.
Collapse
Affiliation(s)
- Juri Murata
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
| | - Yuya Kudo
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
| | - Maho Kotani
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Chiba, Japan (I.K.)
| | - Yoshikane Izawa
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
- Tomita Hospital, Aichi, Japan (Y.T.)
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research (K.F.T.), Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
| | - Kazuto Masamoto
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering (K.M.), University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
41
|
Travasso RDM, Coelho-Santos V. Image-based angio-adaptation modelling: a playground to study cerebrovascular development. Front Physiol 2023; 14:1223308. [PMID: 37565149 PMCID: PMC10411953 DOI: 10.3389/fphys.2023.1223308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Affiliation(s)
- Rui D. M. Travasso
- Department of Physics, Center for Physics of the University of Coimbra (CFisUC), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
| | - Vanessa Coelho-Santos
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
42
|
Crouch EE, Joseph T, Marsan E, Huang EJ. Disentangling brain vasculature in neurogenesis and neurodegeneration using single-cell transcriptomics. Trends Neurosci 2023; 46:551-565. [PMID: 37210315 PMCID: PMC10560453 DOI: 10.1016/j.tins.2023.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/15/2023] [Accepted: 04/26/2023] [Indexed: 05/22/2023]
Abstract
The vasculature is increasingly recognized to impact brain function in health and disease across the life span. During embryonic brain development, angiogenesis and neurogenesis are tightly coupled, coordinating the proliferation, differentiation, and migration of neural and glial progenitors. In the adult brain, neurovascular interactions continue to play essential roles in maintaining brain function and homeostasis. This review focuses on recent advances that leverage single-cell transcriptomics of vascular cells to uncover their subtypes, their organization and zonation in the embryonic and adult brain, and how dysfunction in neurovascular and gliovascular interactions contributes to the pathogenesis of neurodegenerative diseases. Finally, we highlight key challenges for future research in neurovascular biology.
Collapse
Affiliation(s)
- Elizabeth E Crouch
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Tara Joseph
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Elise Marsan
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric J Huang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA; Pathology Service (113B), San Francisco Veterans Administration Health Care System, San Francisco, CA 94121, USA.
| |
Collapse
|
43
|
Wang Y, Du W, Sun Y, Zhang J, Ma C, Jin X. CRTC1 is a potential target to delay aging-induced cognitive deficit by protecting the integrity of the blood-brain barrier via inhibiting inflammation. J Cereb Blood Flow Metab 2023; 43:1042-1059. [PMID: 37086081 PMCID: PMC10291461 DOI: 10.1177/0271678x231169133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
Aging can cause attenuation in the functioning of multiple organs, and blood-brain barrier (BBB) breakdown could promote the occurrence of disorders of the central nervous system during aging. Since inflammation is considered to be an important factor underlying BBB injury during aging, vascular endothelial cell senescence serves as a critical pathological basis for the destruction of BBB integrity. In the current review, we have first introduced the concepts related to aging-induced cognitive deficit and BBB integrity damage. Thereafter, we reviewed the potential relationship between disruption of BBB integrity and cognition deficit and the role of inflammation, vascular endothelial cell senescence, and BBB injury. We have also briefly introduced the function of CREB-regulated transcription co-activator 1 (CRTC1) in cognition and aging-induced CRTC1 changes as well as the critical roles of CRTC1/cyclooxygenase-2 (COX-2) in regulating inflammation, endothelial cell senescence, and BBB injury. Finally, the underlying mechanisms have been summarized and we propose that CRTC1 could be a promising target to delay aging-induced cognitive deficit by protecting the integrity of BBB through promoting inhibition of inflammation-mediated endothelial cell senescence.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurology, the Second Hospital of Jiaxing City, Jiaxing, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yanyun Sun
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfang Zhang
- Department of Physiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Chaolin Ma
- School of Life Science and Institute of Life Science, Nanchang University, Nanchang, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
44
|
Bennett HC, Zhang Q, Wu YT, Chon U, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541998. [PMID: 37305850 PMCID: PMC10257218 DOI: 10.1101/2023.05.23.541998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Aging is the largest risk factor for neurodegenerative disorders, and commonly associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts the vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods (serial two-photon tomography and light sheet microscopy) and in vivo imaging (wide field optical spectroscopy and two-photon imaging) to determine detailed changes in aged cerebrovascular networks. Whole-brain vascular tracing showed an overall ~10% decrease in vascular length and branching density, and light sheet imaging with 3D immunolabeling revealed increased arteriole tortuosity in aged brains. Vasculature and pericyte densities showed significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. Moreover, in vivo imaging in awake mice identified delays in neurovascular coupling and disrupted blood oxygenation. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Equal contribution
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Equal contribution
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Lead contact
| |
Collapse
|
45
|
Qiu B, Zhao Z, Wang N, Feng Z, Chen XJ, Chen W, Sun W, Ge WP, Wang Y. A systematic observation of vasodynamics from different segments along the cerebral vasculature in the penumbra zone of awake mice following cerebral ischemia and recanalization. J Cereb Blood Flow Metab 2023; 43:665-679. [PMID: 36524693 PMCID: PMC10108196 DOI: 10.1177/0271678x221146128] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022]
Abstract
Different segments of the cerebral vascular network may react distinctly to brain ischemia and recanalization. However, there are limited systematic observations of these vascular responses in mice under a physiological state following ischemic stroke. Herein, we aimed to investigate the vasodynamics among several segments along the cerebral vessels in awake mice following cerebral ischemia/recanalization via two-photon imaging. Plasma in the blood vessels were labelled with fluorescein isothiocyanate dextran. Smooth muscle cells and pericytes were labelled via a genetic mouse line (PDGFRβ-tdTomato). We observed a no-reflow phenomenon in downstream microcirculation, and the vasodynamics of different segments of larger cerebral vessels varied in the penumbra area following cerebral ischemia-reperfusion. Despite obtaining reperfusion from the middle cerebral artery, there were significant constrictions of the downstream blood vessels in the ischemic penumbra zone. Interestingly, we observed an extensive constriction of the capillaries 3 hours following recanalization, both at the site covered by pericyte soma and by the pericyte process alone. In addition, we did not observe a significant positive correlation between the changed capillary diameter and pericyte coverage along the capillary. Taken together, abnormal constrictions and vasodynamics of cerebral large and small vessels may directly contribute to microcirculation failure following recanalization in ischemic stroke.
Collapse
Affiliation(s)
- Baoshan Qiu
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
| | - Zichen Zhao
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
| | - Nan Wang
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
| | - Ziyan Feng
- Chinese Institute for Brain Research,
Beijing, China
| | - Xing-jun Chen
- Chinese Institute for Brain Research,
Beijing, China
- Academy for Advanced Interdisciplinary
Studies (AAIS), Peking University, Beijing, China
| | - Weiqi Chen
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Wenzhi Sun
- Chinese Institute for Brain Research,
Beijing, China
- School of Basic Medical Sciences, Capital
Medical University, Beijing, China
| | - Woo-ping Ge
- Chinese Institute for Brain Research,
Beijing, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
- China National Clinical Research Centre for
Neurological Diseases, Beijing, China
- Advanced Innovation Centre for Human Brain
Protection, Capital Medical University, Beijing, China
- National Centre for Neurological Diseases,
Beijing, China
| |
Collapse
|
46
|
Stamenkovic S, Li Y, Waters J, Shih A. Deep Imaging to Dissect Microvascular Contributions to White Matter Degeneration in Rodent Models of Dementia. Stroke 2023; 54:1403-1415. [PMID: 37094035 PMCID: PMC10460612 DOI: 10.1161/strokeaha.122.037156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The increasing socio-economic burden of Alzheimer disease (AD) and AD-related dementias has created a pressing need to define targets for therapeutic intervention. Deficits in cerebral blood flow and neurovascular function have emerged as early contributors to disease progression. However, the cause, progression, and consequence of small vessel disease in AD/AD-related dementias remains poorly understood, making therapeutic targets difficult to pinpoint. Animal models that recapitulate features of AD/AD-related dementias may provide mechanistic insight because microvascular pathology can be studied as it develops in vivo. Recent advances in in vivo optical and ultrasound-based imaging of the rodent brain facilitate this goal by providing access to deeper brain structures, including white matter and hippocampus, which are more vulnerable to injury during cerebrovascular disease. Here, we highlight these novel imaging approaches and discuss their potential for improving our understanding of vascular contributions to AD/AD-related dementias.
Collapse
Affiliation(s)
- Stefan Stamenkovic
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yuandong Li
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Andy Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
47
|
Terman D. Modeling the response of homogeneous and heterogeneous cerebral capillary networks to local changes in vessel diameters. J Theor Biol 2023; 568:111509. [PMID: 37120132 DOI: 10.1016/j.jtbi.2023.111509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023]
Abstract
While microvascular cerebral capillary networks are known to be highly heterogeneous, previous computational models have predicted that heterogeneous cerebral capillary flow patterns result in lower brain tissue partial oxygen pressures. Moreover, as blood flow increases, the flux among capillaries homogenizes. This homogenization of flow is expected to improve the efficiency of oxygenation extraction from the blood. In this work, we use mathematical modeling to explore a possible functional role for the high degree of heterogeneity observed in cerebral capillary networks. Our results suggest that heterogeneity allows for a greater response of tissue oxygen levels to local changes in vessel diameters due to neuronal activation. This result is confirmed for a full 3-dimensional model of capillary networks that includes oxygen diffusion within the tissue region and a reduced model that accounts for changes in capillary blood flow.
Collapse
Affiliation(s)
- David Terman
- Department of Mathematics, The Ohio State University, Columbus, Ohio, 43210 USA.
| |
Collapse
|
48
|
Xia M, Jiao L, Wang XH, Tong M, Yao MD, Li XM, Yao J, Li D, Zhao PQ, Yan B. Single-cell RNA sequencing reveals a unique pericyte type associated with capillary dysfunction. Theranostics 2023; 13:2515-2530. [PMID: 37215579 PMCID: PMC10196835 DOI: 10.7150/thno.83532] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Background: Capillary dysfunction has been implicated in a series of life- threatening vascular diseases characterized by pericyte and endothelial cell (EC) degeneration. However, the molecular profiles that govern the heterogeneity of pericytes have not been fully elucidated. Methods: Single-cell RNA sequencing was conducted on oxygen-induced proliferative retinopathy (OIR) model. Bioinformatics analysis was conducted to identify specific pericytes involved in capillary dysfunction. qRT-PCRs and western blots were conducted to detect Col1a1 expression pattern during capillary dysfunction. Matrigel co-culture assays, PI staining, and JC-1 staining was conducted to determine the role of Col1a1 in pericyte biology. IB4 and NG2 staining was conducted to determine the role of Col1a1 in capillary dysfunction. Results: We constructed an atlas of > 76,000 single-cell transcriptomes from 4 mouse retinas, which could be annotated to 10 distinct retinal cell types. Using the sub-clustering analysis, we further characterized retinal pericytes into 3 different subpopulations. Notably, GO and KEGG pathway analysis demonstrated that pericyte sub-population 2 was identified to be vulnerable to retinal capillary dysfunction. Based on the single-cell sequencing results, Col1a1 was identified as a marker gene of pericyte sub-population 2 and a promising therapeutic target for capillary dysfunction. Col1a1 was abundantly expressed in pericytes and its expression was obviously upregulated in OIR retinas. Col1a1 silencing could retard the recruitment of pericytes toward endothelial cells and aggravated hypoxia-induced pericyte apoptosis in vitro. Col1a1 silencing could reduce the size of neovascular area and avascular area in OIR retinas and suppressed pericyte-myofibroblast transition and endothelial-mesenchymal transition. Moreover, Col1a1 expression was up-regulated in the aqueous humor of the patients with proliferative diabetic retinopathy (PDR) or retinopathy of prematurity (ROP) and up-regulated in the proliferative membranes of PDR patients. Conclusions: These findings enhance the understanding of the complexity and heterogeneity of retinal cells and have important implications for future treatment of capillary dysfunction.
Collapse
Affiliation(s)
- Min Xia
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Lyu Jiao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiao-Han Wang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Min Tong
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Mu-Di Yao
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Xiu-Miao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Jin Yao
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Dan Li
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Pei-Quan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Biao Yan
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200030, China
| |
Collapse
|
49
|
Keep RF, Jones HC, Hamilton MG, Drewes LR. A year in review: brain barriers and brain fluids research in 2022. Fluids Barriers CNS 2023; 20:30. [PMID: 37085841 PMCID: PMC10120509 DOI: 10.1186/s12987-023-00429-0] [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: 04/07/2023] [Indexed: 04/23/2023] Open
Abstract
This aim of this editorial is to highlight progress made in brain barrier and brain fluid research in 2022. It covers studies on the blood-brain, blood-retina and blood-CSF barriers (choroid plexus and meninges), signaling within the neurovascular unit and elements of the brain fluid systems. It further discusses how brain barriers and brain fluid systems are impacted in CNS diseases, their role in disease progression and progress being made in treating such diseases.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | | | - Mark G Hamilton
- Department of Clinical Neurosciences, Division of Neurosurgery, University of Calgary, Alberta, Canada
| | - Lester R Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN, 55812, USA
| |
Collapse
|
50
|
Schreiber S, Bernal J, Arndt P, Schreiber F, Müller P, Morton L, Braun-Dullaeus RC, Valdés-Hernández MDC, Duarte R, Wardlaw JM, Meuth SG, Mietzner G, Vielhaber S, Dunay IR, Dityatev A, Jandke S, Mattern H. Brain Vascular Health in ALS Is Mediated through Motor Cortex Microvascular Integrity. Cells 2023; 12:957. [PMID: 36980297 PMCID: PMC10047140 DOI: 10.3390/cells12060957] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Brain vascular health appears to be critical for preventing the development of amyotrophic lateral sclerosis (ALS) and slowing its progression. ALS patients often demonstrate cardiovascular risk factors and commonly suffer from cerebrovascular disease, with evidence of pathological alterations in their small cerebral blood vessels. Impaired vascular brain health has detrimental effects on motor neurons: vascular endothelial growth factor levels are lowered in ALS, which can compromise endothelial cell formation and the integrity of the blood-brain barrier. Increased turnover of neurovascular unit cells precedes their senescence, which, together with pericyte alterations, further fosters the failure of toxic metabolite removal. We here provide a comprehensive overview of the pathogenesis of impaired brain vascular health in ALS and how novel magnetic resonance imaging techniques can aid its detection. In particular, we discuss vascular patterns of blood supply to the motor cortex with the number of branches from the anterior and middle cerebral arteries acting as a novel marker of resistance and resilience against downstream effects of vascular risk and events in ALS. We outline how certain interventions adapted to patient needs and capabilities have the potential to mechanistically target the brain microvasculature towards favorable motor cortex blood supply patterns. Through this strategy, we aim to guide novel approaches to ALS management and a better understanding of ALS pathophysiology.
Collapse
Affiliation(s)
- Stefanie Schreiber
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Jose Bernal
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
| | - Philipp Arndt
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
| | - Frank Schreiber
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
| | - Patrick Müller
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
- Department of Internal Medicine/Cardiology and Angiology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Lorena Morton
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | | | | | - Roberto Duarte
- Centre for Clinical Brain Sciences, The University of Edinburgh, UK Dementia Research Institute Centre, Edinburgh EH16 4UX, UK
| | - Joanna Marguerite Wardlaw
- Centre for Clinical Brain Sciences, The University of Edinburgh, UK Dementia Research Institute Centre, Edinburgh EH16 4UX, UK
| | - Sven Günther Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Grazia Mietzner
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Ildiko Rita Dunay
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Solveig Jandke
- Department of Neurology, Otto von Guericke University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Department of Biomedical Magnetic Resonance, Faculty of Natural Sciences, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|