1
|
Pushpam M, Talukdar A, Anilkumar S, Maurya SK, Issac TG, Diwakar L. Recurrent endothelin-1 mediated vascular insult leads to cognitive impairment protected by trophic factor pleiotrophin. Exp Neurol 2024; 381:114938. [PMID: 39197707 DOI: 10.1016/j.expneurol.2024.114938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/12/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
Vascular dementia (VaD) is a complex neurodegenerative condition, with cerebral small vessel dysfunctions as the central role in its pathogenesis. Given the lack of suitable animal models to study the disease pathogenesis, we developed a mouse model to closely emulate the clinical scenarios of recurrent transient ischemic attacks (TIAs) leading to VaD using vasoconstricting peptide Endothelin-1(ET-1). We observed that administration of ET-1 led to blood-brain barrier (BBB) disruption and detrimental changes in its components, such as endothelial cells and pericytes, along with neuronal loss and synaptic dysfunction, resulting in irreversible memory loss. Further, in our pursuit of understanding potential interventions, we co-administered pleiotrophin (PTN) alongside ET-1 injections. PTN exhibited remarkable efficacy in preserving vital components of the BBB, including endothelial cells and pericytes, thereby restoring BBB integrity, preventing neuronal loss, and enhancing memory function. Our findings give a valuable framework for understanding the detrimental effects of multiple TIAs on brain health and provide a useful animal model to explore VaD's underlying mechanisms further and pave the way for promising therapies.
Collapse
Affiliation(s)
- Mayank Pushpam
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India; Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Ankita Talukdar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Shobha Anilkumar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | | | - Thomas Gregor Issac
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
2
|
McCloskey MC, Ahmad SD, Widom LP, Kasap P, Gastfriend BD, Shusta EV, Palecek SP, Engelhardt B, Gaborski TR, Flax J, Waugh RE, McGrath JL. Pericytes Enrich the Basement Membrane and Reduce Neutrophil Transmigration in an In Vitro Model of Peripheral Inflammation at the Blood-Brain Barrier. Biomater Res 2024; 28:0081. [PMID: 39363889 PMCID: PMC11447289 DOI: 10.34133/bmr.0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 10/05/2024] Open
Abstract
Sepsis is the most lethal and expensive condition treated in intensive care units. Sepsis survivors frequently suffer long-term cognitive impairment, which has been linked to the breakdown of the blood-brain barrier (BBB) during a sepsis-associated "cytokine storm". Because animal models poorly recapitulate sepsis pathophysiology, human models are needed to understand sepsis-associated brain injury and to develop novel therapeutic strategies. With the concurrent emergence of tissue chip technologies and the maturation of protocols for human induced pluripotent stem cell (hiPSC), we can now develop advanced in vitro models of the human BBB and immune system to understand the relationship between systemic inflammation and brain injury. Here, we present a BBB model of the primary barrier developed on the μSiM (microphysiological system enabled by an ultrathin silicon nanomembrane) tissue chip platform. The model features isogenically matched hiPSC-derived extended endothelial culture method brain microvascular endothelial cell-like cells (EECM-BMEC-like cells) and brain pericyte-like cells (BPLCs) in a back-to-back coculture separated by the ultrathin (100 nm) membrane. Both endothelial monocultures and cocultures with pericytes responded to sepsis-like stimuli, with increased small-molecule permeability, although no differences were detected between culture conditions. Conversely, BPLC coculture reduced the number of neutrophils that crossed the EECM-BMEC-like cell monolayer under sepsis-like stimulation. Interestingly, this barrier protection was not seen when the stimulus originated from the tissue side. Our studies are consistent with the reported role for pericytes in regulating leukocyte trafficking during sepsis but indicate that EECM-BMEC-like cells alone are sufficient to maintain the restrictive small-molecule permeability of the BBB.
Collapse
Affiliation(s)
- Molly C. McCloskey
- Department of Biomedical Engineering,
University of Rochester, Rochester NY, USA
| | - S. Danial Ahmad
- Department of Biomedical Engineering,
University of Rochester, Rochester NY, USA
| | - Louis P. Widom
- Department of Biomedical Engineering,
Rochester Institute of Technology, Rochester NY, USA
| | - Pelin Kasap
- Theodor Kocher Institute,
University of Bern, Bern, Switzerland
| | - Benjamin D. Gastfriend
- Department of Chemical and Biological Engineering,
University of Wisconsin–Madison, Madison, WI, USA
- Departments of Pharmacology and Neurosciences,
University of California, San Diego, La Jolla, CA, USA
| | - Eric V. Shusta
- Department of Chemical and Biological Engineering,
University of Wisconsin–Madison, Madison, WI, USA
- Department of Neurological Surgery,
University of Wisconsin–Madison, Madison, WI, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering,
University of Wisconsin–Madison, Madison, WI, USA
| | | | - Thomas R. Gaborski
- Department of Biomedical Engineering,
Rochester Institute of Technology, Rochester NY, USA
| | - Jonathan Flax
- Department of Biomedical Engineering,
University of Rochester, Rochester NY, USA
| | - Richard E. Waugh
- Department of Biomedical Engineering,
University of Rochester, Rochester NY, USA
| | - James L. McGrath
- Department of Biomedical Engineering,
University of Rochester, Rochester NY, USA
| |
Collapse
|
3
|
Zielniok K, Rusinek K, Słysz A, Lachota M, Bączyńska E, Wiewiórska-Krata N, Szpakowska A, Ciepielak M, Foroncewicz B, Mucha K, Zagożdżon R, Pojda Z. 3D-Bioprinted Co-Cultures of Glioblastoma Multiforme and Mesenchymal Stromal Cells Indicate a Role for Perivascular Niche Cells in Shaping Glioma Chemokine Microenvironment. Cells 2024; 13:1404. [PMID: 39272976 PMCID: PMC11393941 DOI: 10.3390/cells13171404] [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: 07/10/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
3D bioprinting has become a valuable tool for studying the biology of solid tumors, including glioblastoma multiforme (GBM). Our analysis of publicly available bulk RNA and single-cell sequencing data has allowed us to define the chemotactic profile of GBM tumors and identify the cell types that secrete particular chemokines in the GBM tumor microenvironment (TME). Our findings indicate that primary GBM tissues express multiple chemokines, whereas spherical monocultures of GBM cells significantly lose this diversity. Subsequently, the comparative analysis of GBM spherical monocultures vs. 3D-bioprinted multicultures of cells showed a restoration of chemokine profile diversity in 3D-bioprinted cultures. Furthermore, single-cell RNA-Seq analysis showed that cells of the perivascular niche (pericytes and endocytes) express multiple chemokines in the GBM TME. Next, we 3D-bioprinted cells from two glioblastoma cell lines, U-251 and DK-MG, alone and as co-cultures with mesenchymal stromal cells (representing cells of the perivascular niche) and assessed the chemokine secretome. The results clearly demonstrated that the interaction of tumors and mesenchymal cells leads to in a significant increase in the repertoire and levels of secreted chemokines under culture in 21% O2 and 1% O2. Our study indicates that cells of the perivascular niche may perform a substantial role in shaping the chemokine microenvironment in GBM tumors.
Collapse
Affiliation(s)
- Katarzyna Zielniok
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Kinga Rusinek
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Anna Słysz
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Mieszko Lachota
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Ewa Bączyńska
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Natalia Wiewiórska-Krata
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Promix (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Anna Szpakowska
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Martyna Ciepielak
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Bartosz Foroncewicz
- Promix (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Krzysztof Mucha
- Promix (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, 02-006 Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Radosław Zagożdżon
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Zygmunt Pojda
- Department of Regenerative Medicine, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
4
|
Öztürk M, Turgut FS, Akbalık D, Demirkıran ME, Kaplan İ. Serum Erythropoietin and Ischemic-Modified Albumin Levels in Adolescents with Obsessive-Compulsive Disorder. J Mol Neurosci 2024; 74:67. [PMID: 38995319 PMCID: PMC11245444 DOI: 10.1007/s12031-024-02247-x] [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: 05/23/2024] [Accepted: 07/09/2024] [Indexed: 07/13/2024]
Abstract
Erythropoietin (EPO) has neuroprotective effects by increasing oxidative stress resistance and stabilizing redox balance. Ischemic-modified albumin (IMA) is a product of protein oxidation, and recent evidence suggests that IMA can be used as an indicator of oxidative damage. This study aimed to investigate serum EPO and IMA levels in obsessive-compulsive disorder (OCD) patients and to investigate the relationship between EPO and IMA levels and clinical variables such as disease duration and disease severity. A total of 68 adolescents (11-18 years old), including 35 OCD patients (18 males/17 females) and 33 healthy controls (14 males/19 females) without comorbid disorders matched for age, gender, and BMI, were included in the study. The enzyme-amplified chemiluminescence technique determined serum EPO levels, and serum IMA levels were determined by the spectrophotometric method. Serum EPO levels were lower in OCD patients compared to healthy controls (p = 0.002; Z = - 3.123), and serum IMA levels (ABSU) were significantly higher in the OCD group (p = 0.005). A significant positive correlation was found between IMA levels and the duration of OCD symptoms (p = 0.015, r = 0.409). The study's findings contribute to the growing body of evidence implicating inflammatory and oxidative processes in the pathogenesis of OCD. The potential of EPO and IMA levels as diagnostic biomarkers for OCD aligns with the ongoing efforts to identify reliable biological markers for the disorder. The positive correlation of IMA levels with the duration of OCD shows the importance of early detection of oxidative damage.
Collapse
Affiliation(s)
- Masum Öztürk
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dicle University, Diyarbakır, Turkey.
| | - Fatma Subaşı Turgut
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Davut Akbalık
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Mustafa Erhan Demirkıran
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - İbrahim Kaplan
- Department of Biochemistry, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| |
Collapse
|
5
|
Whitehead B, Corbin D, Albowaidey A, Zhang N, Karelina K, Weil ZM. Mild traumatic brain injury induces pericyte detachment independent of stroke vulnerability. Neurosci Lett 2024; 818:137552. [PMID: 37949292 PMCID: PMC10913758 DOI: 10.1016/j.neulet.2023.137552] [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: 09/12/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Mild traumatic brain injury (mTBI) is an independent risk factor for ischemic stroke and can result in poorer outcomes- an effect presumed to involve the cerebral vasculature. Here we tested the hypothesis that mTBI-induced pericyte detachment from the cerebrovascular endothelium is responsible for worsened stroke outcomes. We performed a mild closed-head injury and/or treated C57/bl6 mice with imatinib mesylate, a tyrosine kinase inhibitor that induces pericyte detachment. The time course of pericyte detachment was assessed 7, 14, and 28 days post injury (DPI). To test the role of pericytes in TBI-induced exacerbation of ischemic stroke outcomes, we induced mTBI and/or treated mice with imatinib for one week prior to transient middle cerebral artery occlusion. We found that injury promoted pericyte detachment from the vasculature commensurate with the levels of detachment seen in imatinib-only treated animals, and that the detachment persisted for at least 14DPI, but recovered to sham levels by 28DPI. Further, mTBI, but not imatinib-induced pericyte detachment, increased infarct volume. Thus, we conclude that the transient detachment of pericytes caused by mTBI may not be sufficient to exacerbate subsequent ischemic stroke damage. These data have important implications for understanding cerebrovascular dysfunction following mTBI and potential mechanisms of increased risk for future ischemic strokes.
Collapse
Affiliation(s)
- Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA.
| | - Deborah Corbin
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Ali Albowaidey
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Zachary M Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| |
Collapse
|
6
|
Whitehead B, Karelina K, Weil ZM. Pericyte dysfunction is a key mediator of the risk of cerebral ischemia. J Neurosci Res 2023; 101:1840-1848. [PMID: 37724604 DOI: 10.1002/jnr.25245] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/15/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
Pericytes are critical yet understudied cells that are a central component of the neurovascular unit. They are connected to the cerebrovascular endothelium and help control vascular contractility and maintain the blood-brain barrier. Pericyte dysfunction has the potential to mediate many of the deleterious vascular consequences of ischemic stroke. Current therapeutics are designed to be administered after stroke onset and limit damage, but there are few options to target vascular risk factors pre-ischemia which likely contribute to stroke outcomes. Here, we focus on the role of pericytes in health and disease, and discuss how pericyte dysfunction can increase the risk of ischemic injury. Additionally, we note that despite the importance of pericytes in cerebrovascular disease, there are relatively few current therapeutic options that target pericyte function.
Collapse
Affiliation(s)
- Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Zachary M Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
7
|
Huang S, de Jong D, Das JP, Widemon RS, Braumuller B, Paily J, Deng A, Liou C, Roa T, Huang A, Ma H, D'Souza B, Leb J, L'Hereaux J, Nguyen P, Luk L, Francescone M, Yeh R, Maccarrone V, Dercle L, Salvatore MM, Capaccione KM. Imaging the Side Effects of CAR T Cell Therapy: A Primer for the Practicing Radiologist. Acad Radiol 2023; 30:2712-2727. [PMID: 37394411 DOI: 10.1016/j.acra.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 07/04/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a revolutionary form of immunotherapy that has proven to be efficacious in the treatment of many hematologic cancers. CARs are modified T lymphocytes that express an artificial receptor specific to a tumor-associated antigen. These engineered cells are then reintroduced to upregulate the host immune responses and eradicate malignant cells. While the use of CAR T cell therapy is rapidly expanding, little is known about how common side effects such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity (ICANS) present radiographically. Here we provide a comprehensive review of how side effects present in different organ systems and how they can be optimally imaged. Early and accurate recognition of the radiographic presentation of these side effects is critical to the practicing radiologist and their patients so that these side effects can be promptly identified and treated.
Collapse
Affiliation(s)
- Sophia Huang
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Dorine de Jong
- Department of Immunology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 (D.J.)
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 (J.D., R.Y.)
| | - Reginald Scott Widemon
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Brian Braumuller
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Jacienta Paily
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Aileen Deng
- Department of Hematology and Oncology, Novant Health, 170 Medical Park Road, Mooresville, North Carolina 28117 (A.D.)
| | - Connie Liou
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Tina Roa
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Alice Huang
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Jade L'Hereaux
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Pamela Nguyen
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Mark Francescone
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 (J.D., R.Y.)
| | - Valerie Maccarrone
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Mary M Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.)
| | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168th Street, New York, New York 10032 (S.H., R.S.W., B.B., J.P., C.L., T.R., A.H., H.M., B.D.S., J.L., J.L.H., P.N., L.L., M.F., V.M., L.D., M.S., K.M.C.).
| |
Collapse
|
8
|
Rios-Hoyo A, Arriola E. Immunotherapy and brain metastasis in lung cancer: connecting bench side science to the clinic. Front Immunol 2023; 14:1221097. [PMID: 37876939 PMCID: PMC10590916 DOI: 10.3389/fimmu.2023.1221097] [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: 05/11/2023] [Accepted: 09/15/2023] [Indexed: 10/26/2023] Open
Abstract
Brain metastases (BMs) are the most common form of intracranial malignant neoplasms in adults, with a profound impact on quality of life and traditionally associated with a dismal prognosis. Lung cancer accounts for approximately 40%-50% of BM across different tumors. The process leading to BMs is complex and includes local invasion, intravasation, tumor cells circulation into the bloodstream, disruption of the blood-brain barrier, extravasation of tumor cells into the brain parenchyma, and interaction with cells of the brain microenvironment, among others. Once the tumor cells have seeded in the brain parenchyma, they encounter different glial cells of the brain, as well as immune cells. The interaction between these cells and tumor cells is complex and is associated with both antitumoral and protumoral effects. To overcome the lethal prognosis associated with BMs, different treatment strategies have been developed, such as immunotherapy with immune checkpoint inhibitors, particularly inhibitors of the PD-1/PD-L1 axis, which have demonstrated to be an effective treatment in both non-small cell lung cancer and small cell lung cancer. These antibodies have shown to be effective in the treatment of BM, alone or in combination with chemotherapy or radiotherapy. However, many unsolved questions remain to be answered, such as the sequencing of immunotherapy and radiotherapy, the optimal management in symptomatic BMs, the role of the addition of anti-CTLA-4 antibodies, and so forth. The complexity in the management of BMs in the era of immunotherapy requires a multidisciplinary approach to adequately treat this devastating event. The aim of this review is to summarize evidence regarding epidemiology of BM, its pathophysiology, current approach to treatment strategies, as well as future perspectives.
Collapse
Affiliation(s)
- Alejandro Rios-Hoyo
- Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Edurne Arriola
- Department of Medical Oncology, Hospital del Mar-CIBERONC (Centro de Investigación Biomédica en Red de Oncología), Barcelona, Spain
- Cancer Research Program, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
| |
Collapse
|
9
|
Benarroch E. What Are the Roles of Pericytes in the Neurovascular Unit and Its Disorders? Neurology 2023; 100:970-977. [PMID: 37188542 PMCID: PMC10186232 DOI: 10.1212/wnl.0000000000207379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 05/17/2023] Open
|
10
|
Şekerdağ-Kılıç E, Ulusoy C, Atak D, Özkan E, Gökyüzü AB, Seyaj S, Deniz G, Uçar EA, Budan AS, Zeybel M, Öztop-Çakmak Ö, Vural A, Tuncer A, Karabudak R, Kücükali CI, Tüzün E, Gürsoy-Özdemir Y. Perivascular PDGFRB+ cells accompany lesion formation and clinical evolution differentially in two different EAE models. Mult Scler Relat Disord 2023; 69:104428. [PMID: 36450174 DOI: 10.1016/j.msard.2022.104428] [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: 08/01/2022] [Revised: 10/31/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Multiple Sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that may lead to progressive disability. Here, we explored the behavioral pattern and the role of vasculature especially PDGFRB+ pericytes/ perivascular cells, in MS pathogenesis. METHODS We have evaluated vascular changes in two different experimental allergic encephalomyelitis (EAE) mice models (MOG and PLP-induced). PDGFRB+ cells demonstrated distinct and different behavioral patterns. In both models, fibrosis formation was detected via collagen, fibronectin, and extracellular matrix accumulation. RESULTS The PLP-induced animal model revealed that fibrosis predominantly occurs in perivascular locations and that PDGFRB+ cells are accumulated around vessels. Also, the expression of fibrotic genes and genes coding extracellular matrix (ECM) proteins are upregulated. Moreover, the perivascular thick wall structures in affected vessels of this model presented primarily increased PDGFRB+ cells but not NG2+ cells in the transgenic NG2-DsRed transgenic animal model. On the other hand, in MOG induced model, PDGFRB+ perivascular cells were accumulated at the lesion sites. PDGFRB+ cells colocalized with ECM proteins (collagen, fibronectin, and lysyl oxidase L3). Nevertheless, both MOG and PLP-immunized mice showed increasing EAE severity, and disability parallel with enhanced perivascular cell accumulation as the disease progressed from earlier (day 15) to later (day 40). CONCLUSION As a result, we have concluded that PDGFRB+ perivascular cells may be participating in lesion progression and as well as demonstrating different responses in different EAE models.
Collapse
Affiliation(s)
- Emine Şekerdağ-Kılıç
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Canan Ulusoy
- Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Dila Atak
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Esra Özkan
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Aysu Bilge Gökyüzü
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Seddiq Seyaj
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Gülsüm Deniz
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Ege Anil Uçar
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Abdullah Salih Budan
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey
| | - Müjdat Zeybel
- Department of Gastroenterology and Hepatology, School of Medicine, Koҫ University, Istanbul, Turkey
| | - Özgür Öztop-Çakmak
- Department of Neurology, School of Medicine, Koҫ University, Istanbul, Turkey
| | - Atay Vural
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey; Department of Neurology, School of Medicine, Koҫ University, Istanbul, Turkey
| | - Asli Tuncer
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Rana Karabudak
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Erdem Tüzün
- Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Turkey; Department of Neurology, School of Medicine, Koҫ University, Istanbul, Turkey.
| |
Collapse
|
11
|
He H, Yang W, Su N, Zhang C, Dai J, Han F, Singhal M, Bai W, Zhu X, Zhu J, Liu Z, Xia W, Liu X, Zhang C, Jiang K, Huang W, Chen D, Wang Z, He X, Kirchhoff F, Li Z, Liu C, Huan J, Wang X, Wei W, Wang J, Augustin HG, Hu J. Activating NO-sGC crosstalk in the mouse vascular niche promotes vascular integrity and mitigates acute lung injury. J Exp Med 2022; 220:213673. [PMID: 36350314 PMCID: PMC9984546 DOI: 10.1084/jem.20211422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/20/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Disruption of endothelial cell (ECs) and pericytes interactions results in vascular leakage in acute lung injury (ALI). However, molecular signals mediating EC-pericyte crosstalk have not been systemically investigated, and whether targeting such crosstalk could be adopted to combat ALI remains elusive. Using comparative genome-wide EC-pericyte crosstalk analysis of healthy and LPS-challenged lungs, we discovered that crosstalk between endothelial nitric oxide and pericyte soluble guanylate cyclase (NO-sGC) is impaired in ALI. Indeed, stimulating the NO-sGC pathway promotes vascular integrity and reduces lung edema and inflammation-induced lung injury, while pericyte-specific sGC knockout abolishes this protective effect. Mechanistically, sGC activation suppresses cytoskeleton rearrangement in pericytes through inhibiting VASP-dependent F-actin formation and MRTFA/SRF-dependent de novo synthesis of genes associated with cytoskeleton rearrangement, thereby leading to the stabilization of EC-pericyte interactions. Collectively, our data demonstrate that impaired NO-sGC crosstalk in the vascular niche results in elevated vascular permeability, and pharmacological activation of this crosstalk represents a promising translational therapy for ALI.
Collapse
Affiliation(s)
- Hao He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Nan Su
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chuankai Zhang
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianing Dai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Feng Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wenjuan Bai
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolan Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Zhen Liu
- University of Chinese Academy of Sciences, Beijing, China,Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoting Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chonghe Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Kai Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Dan Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhaoyin Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Xueyang He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Zhenyu Li
- Texas A&M Health Science Center, Bryan, TX
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wu Wei
- University of Chinese Academy of Sciences, Beijing, China,Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China,Correspondence to Junhao Hu:
| |
Collapse
|
12
|
Mitusova K, Peltek OO, Karpov TE, Muslimov AR, Zyuzin MV, Timin AS. Overcoming the blood-brain barrier for the therapy of malignant brain tumor: current status and prospects of drug delivery approaches. J Nanobiotechnology 2022; 20:412. [PMID: 36109754 PMCID: PMC9479308 DOI: 10.1186/s12951-022-01610-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/18/2022] [Indexed: 01/06/2023] Open
Abstract
Besides the broad development of nanotechnological approaches for cancer diagnosis and therapy, currently, there is no significant progress in the treatment of different types of brain tumors. Therapeutic molecules crossing the blood-brain barrier (BBB) and reaching an appropriate targeting ability remain the key challenges. Many invasive and non-invasive methods, and various types of nanocarriers and their hybrids have been widely explored for brain tumor treatment. However, unfortunately, no crucial clinical translations were observed to date. In particular, chemotherapy and surgery remain the main methods for the therapy of brain tumors. Exploring the mechanisms of the BBB penetration in detail and investigating advanced drug delivery platforms are the key factors that could bring us closer to understanding the development of effective therapy against brain tumors. In this review, we discuss the most relevant aspects of the BBB penetration mechanisms, observing both invasive and non-invasive methods of drug delivery. We also review the recent progress in the development of functional drug delivery platforms, from viruses to cell-based vehicles, for brain tumor therapy. The destructive potential of chemotherapeutic drugs delivered to the brain tumor is also considered. This review then summarizes the existing challenges and future prospects in the use of drug delivery platforms for the treatment of brain tumors.
Collapse
Affiliation(s)
- Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Oleksii O Peltek
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Albert R Muslimov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
- Sirius University of Science and Technology, Olympic Ave 1, Sirius, 354340, Russian Federation
| | - Mikhail V Zyuzin
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation.
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation.
| |
Collapse
|
13
|
Jones CFE, Di Cio S, Connelly JT, Gautrot JE. Design of an Integrated Microvascularized Human Skin-on-a-Chip Tissue Equivalent Model. Front Bioeng Biotechnol 2022; 10:915702. [PMID: 35928950 PMCID: PMC9343775 DOI: 10.3389/fbioe.2022.915702] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue-engineered skin constructs have been under development since the 1980s as a replacement for human skin tissues and animal models for therapeutics and cosmetic testing. These have evolved from simple single-cell assays to increasingly complex models with integrated dermal equivalents and multiple cell types including a dermis, epidermis, and vasculature. The development of micro-engineered platforms and biomaterials has enabled scientists to better recreate and capture the tissue microenvironment in vitro, including the vascularization of tissue models and their integration into microfluidic chips. However, to date, microvascularized human skin equivalents in a microfluidic context have not been reported. Here, we present the design of a novel skin-on-a-chip model integrating human-derived primary and immortalized cells in a full-thickness skin equivalent. The model is housed in a microfluidic device, in which a microvasculature was previously established. We characterize the impact of our chip design on the quality of the microvascular networks formed and evidence that this enables the formation of more homogenous networks. We developed a methodology to harvest tissues from embedded chips, after 14 days of culture, and characterize the impact of culture conditions and vascularization (including with pericyte co-cultures) on the stratification of the epidermis in the resulting skin equivalents. Our results indicate that vascularization enhances stratification and differentiation (thickness, architecture, and expression of terminal differentiation markers such as involucrin and transglutaminase 1), allowing the formation of more mature skin equivalents in microfluidic chips. The skin-on-a-chip tissue equivalents developed, because of their realistic microvasculature, may find applications for testing efficacy and safety of therapeutics delivered systemically, in a human context.
Collapse
Affiliation(s)
- Christian F. E. Jones
- Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Stefania Di Cio
- Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - John T. Connelly
- The Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Julien E. Gautrot
- Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
14
|
Jung O, Thomas A, Burks SR, Dustin ML, Frank JA, Ferrer M, Stride E. Neuroinflammation associated with ultrasound-mediated permeabilization of the blood-brain barrier. Trends Neurosci 2022; 45:459-470. [PMID: 35461727 PMCID: PMC9117477 DOI: 10.1016/j.tins.2022.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/17/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
The blood-brain barrier (BBB) continues to represent one of the most significant challenges for successful drug-based treatments of neurological disease. Mechanical modulation of the BBB using focused ultrasound (FUS) and microbubbles (MBs) has shown considerable promise in enhancing the delivery of therapeutics to the brain, but questions remain regarding possible long-term effects of such forced disruption. This review examines the evidence for inflammation associated with ultrasound-induced BBB disruption and potential strategies for managing such inflammatory effects to improve both the efficacy and safety of therapeutic ultrasound in neurological applications.
Collapse
Affiliation(s)
- Olive Jung
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK; 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Alec Thomas
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Scott R Burks
- The Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Michael L Dustin
- Nuffield Department of Orthopedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Joseph A Frank
- The Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Eleanor Stride
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Rudge JD. A New Hypothesis for Alzheimer's Disease: The Lipid Invasion Model. J Alzheimers Dis Rep 2022; 6:129-161. [PMID: 35530118 PMCID: PMC9028744 DOI: 10.3233/adr-210299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
This paper proposes a new hypothesis for Alzheimer's disease (AD)-the lipid invasion model. It argues that AD results from external influx of free fatty acids (FFAs) and lipid-rich lipoproteins into the brain, following disruption of the blood-brain barrier (BBB). The lipid invasion model explains how the influx of albumin-bound FFAs via a disrupted BBB induces bioenergetic changes and oxidative stress, stimulates microglia-driven neuroinflammation, and causes anterograde amnesia. It also explains how the influx of external lipoproteins, which are much larger and more lipid-rich, especially more cholesterol-rich, than those normally present in the brain, causes endosomal-lysosomal abnormalities and overproduction of the peptide amyloid-β (Aβ). This leads to the formation of amyloid plaques and neurofibrillary tangles, the most well-known hallmarks of AD. The lipid invasion model argues that a key role of the BBB is protecting the brain from external lipid access. It shows how the BBB can be damaged by excess Aβ, as well as by most other known risk factors for AD, including aging, apolipoprotein E4 (APOE4), and lifestyle factors such as hypertension, smoking, obesity, diabetes, chronic sleep deprivation, stress, and head injury. The lipid invasion model gives a new rationale for what we already know about AD, explaining its many associated risk factors and neuropathologies, including some that are less well-accounted for in other explanations of AD. It offers new insights and suggests new ways to prevent, detect, and treat this destructive disease and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan D’Arcy Rudge
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
16
|
Li W, Cao F, Takase H, Arai K, Lo EH, Lok J. Blood-Brain Barrier Mechanisms in Stroke and Trauma. Handb Exp Pharmacol 2022; 273:267-293. [PMID: 33580391 DOI: 10.1007/164_2020_426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The brain microenvironment is tightly regulated. The blood-brain barrier (BBB), which is composed of cerebral endothelial cells, astrocytes, and pericytes, plays an important role in maintaining the brain homeostasis by regulating the transport of both beneficial and detrimental substances between circulating blood and brain parenchyma. After brain injury and disease, BBB tightness becomes dysregulated, thus leading to inflammation and secondary brain damage. In this chapter, we overview the fundamental mechanisms of BBB damage and repair after stroke and traumatic brain injury (TBI). Understanding these mechanisms may lead to therapeutic opportunities for brain injury.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang Cao
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Josephine Lok
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Yuan M, Wang Y, Wang S, Huang Z, Jin F, Zou Q, Li J, Pu Y, Cai Z. Bioenergetic Impairment in the Neuro-Glia-Vascular Unit: An Emerging Physiopathology during Aging. Aging Dis 2021; 12:2080-2095. [PMID: 34881087 PMCID: PMC8612602 DOI: 10.14336/ad.2021.04017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/17/2021] [Indexed: 12/28/2022] Open
Abstract
An emerging concept termed the "neuro-glia-vascular unit" (NGVU) has been established in recent years to understand the complicated mechanism of multicellular interactions among vascular cells, glial cells, and neurons. It has been proverbially reported that the NGVU is significantly associated with neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Physiological aging is an inevitable progression associated with oxidative damage, bioenergetic alterations, mitochondrial dysfunction, and neuroinflammation, which is partially similar to the pathology of AD. Thus, senescence is regarded as the background for the development of neurodegenerative diseases. With the exacerbation of global aging, senescence is an increasingly serious problem in the medical field. In this review, the coupling of each component, including neurons, glial cells, and vascular cells, in the NGVU is described in detail. Then, various mechanisms of age-dependent impairment in each part of the NGVU are discussed. Moreover, the potential bioenergetic alterations between different cell types in the NGVU are highlighted, which seems to be an emerging physiopathology associated with the aged brain. Bioenergetic intervention in the NGVU may be a new direction for studies on delaying or diminishing aging in the future.
Collapse
Affiliation(s)
- Minghao Yuan
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Yangyang Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Shengyuan Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Zhenting Huang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Feng Jin
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Qian Zou
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Jing Li
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Yinshuang Pu
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Zhiyou Cai
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Shaheryar ZA, Khan MA, Adnan CS, Zaidi AA, Hänggi D, Muhammad S. Neuroinflammatory Triangle Presenting Novel Pharmacological Targets for Ischemic Brain Injury. Front Immunol 2021; 12:748663. [PMID: 34691061 PMCID: PMC8529160 DOI: 10.3389/fimmu.2021.748663] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is one of the leading causes of morbidity and mortality globally. Hundreds of clinical trials have proven ineffective in bringing forth a definitive and effective treatment for ischemic stroke, except a myopic class of thrombolytic drugs. That, too, has little to do with treating long-term post-stroke disabilities. These studies proposed diverse options to treat stroke, ranging from neurotropic interpolation to venting antioxidant activity, from blocking specific receptors to obstructing functional capacity of ion channels, and more recently the utilization of neuroprotective substances. However, state of the art knowledge suggests that more pragmatic focus in finding effective therapeutic remedy for stroke might be targeting intricate intracellular signaling pathways of the 'neuroinflammatory triangle': ROS burst, inflammatory cytokines, and BBB disruption. Experimental evidence reviewed here supports the notion that allowing neuroprotective mechanisms to advance, while limiting neuroinflammatory cascades, will help confine post-stroke damage and disabilities.
Collapse
Affiliation(s)
- Zaib A. Shaheryar
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Faculty of Pharmacy, University of Lahore, Lahore, Pakistan
| | - Mahtab A. Khan
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | | | - Awais Ali Zaidi
- Faculty of Pharmacy, University of Lahore, Lahore, Pakistan
- Imran Idrees College of Pharmacy, Lahore, Pakistan
| | - Daniel Hänggi
- Department of Neurosurgery, Faculty of Medicine and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Neurosurgery, Faculty of Medicine and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
19
|
Cerebral Pericytes and Endothelial Cells Communicate through Inflammasome-Dependent Signals. Int J Mol Sci 2021; 22:ijms22116122. [PMID: 34204159 PMCID: PMC8201302 DOI: 10.3390/ijms22116122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/27/2022] Open
Abstract
By upregulation of cell adhesion molecules and secretion of proinflammatory cytokines, cells of the neurovascular unit, including pericytes and endothelial cells, actively participate in neuroinflammatory reactions. As previously shown, both cell types can activate inflammasomes, cerebral endothelial cells (CECs) through the canonical pathway, while pericytes only through the noncanonical pathway. Using complex in vitro models, we demonstrate here that the noncanonical inflammasome pathway can be induced in CECs as well, leading to a further increase in the secretion of active interleukin-1β over that observed in response to activation of the canonical pathway. In parallel, a more pronounced disruption of tight junctions takes place. We also show that CECs respond to inflammatory stimuli coming from both the apical/blood and the basolateral/brain directions. As a result, CECs can detect factors secreted by pericytes in which the noncanonical inflammasome pathway is activated and respond with inflammatory activation and impairment of the barrier properties. In addition, upon sensing inflammatory signals, CECs release inflammatory factors toward both the blood and the brain sides. Consequently, CECs activate pericytes by upregulating their expression of NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3), an inflammasome-forming pattern recognition receptor. In conclusion, cerebral pericytes and endothelial cells mutually activate each other in inflammation.
Collapse
|
20
|
Oliveira-Giacomelli Á, Petiz LL, Andrejew R, Turrini N, Silva JB, Sack U, Ulrich H. Role of P2X7 Receptors in Immune Responses During Neurodegeneration. Front Cell Neurosci 2021; 15:662935. [PMID: 34122013 PMCID: PMC8187565 DOI: 10.3389/fncel.2021.662935] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/26/2021] [Indexed: 01/16/2023] Open
Abstract
P2X7 receptors are ion-gated channels activated by ATP. Under pathological conditions, the extensive release of ATP induces sustained P2X7 receptor activation, culminating in induction of proinflammatory pathways with inflammasome assembly and cytokine release. These inflammatory conditions, whether occurring peripherally or in the central nervous system (CNS), increase blood-brain-barrier (BBB) permeability. Besides its well-known involvement in neurodegeneration and neuroinflammation, the P2X7 receptor may induce BBB disruption and chemotaxis of peripheral immune cells to the CNS, resulting in brain parenchyma infiltration. For instance, despite common effects on cytokine release, P2X7 receptor signaling is also associated with metalloproteinase secretion and activation, as well as migration and differentiation of T lymphocytes, monocytes and dendritic cells. Here we highlight that peripheral immune cells mediate the pathogenesis of Multiple Sclerosis and Parkinson's and Alzheimer's disease, mainly through T lymphocyte, neutrophil and monocyte infiltration. We propose that P2X7 receptor activation contributes to neurodegenerative disease progression beyond its known effects on the CNS. This review discusses how P2X7 receptor activation mediates responses of peripheral immune cells within the inflamed CNS, as occurring in the aforementioned diseases.
Collapse
Affiliation(s)
| | - Lyvia Lintzmaier Petiz
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Roberta Andrejew
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Natalia Turrini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Jean Bezerra Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Ulrich Sack
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
21
|
Choi YK, Kim YM. Regulation of Endothelial and Vascular Functions by Carbon Monoxide via Crosstalk With Nitric Oxide. Front Cardiovasc Med 2021; 8:649630. [PMID: 33912601 PMCID: PMC8071856 DOI: 10.3389/fcvm.2021.649630] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Carbon monoxide (CO), generated by heme oxygenase (HO), has been considered a signaling molecule in both the cardiovascular and central nervous systems. The biological function of the HO/CO axis is mostly related to other gaseous molecules, including nitric oxide (NO), which is synthesized by nitric oxide synthase (NOS). Healthy blood vessels are essential for the maintenance of tissue homeostasis and whole-body metabolism; however, decreased or impaired vascular function is a high-risk factor of cardiovascular and neuronal diseases. Accumulating evidence supports that the interplay between CO and NO plays a crucial role in vascular homeostasis and regeneration by improving endothelial function. Moreover, endothelial cells communicate with neighboring cells, such as, smooth muscle cells, immune cells, pericytes, and astrocytes in the periphery and neuronal vascular systems. Endogenous CO could mediate the cell-cell communication and improve the physiological functions of the cardiovascular and neurovascular systems via crosstalk with NO. Thus, a forward, positive feedback circuit between HO/CO and NOS/NO pathways can maintain cardiovascular and neurovascular homeostasis and prevent various human diseases. We discussed the crucial role of CO-NO crosstalk in the cardiovascular and neurovascular systems.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
22
|
Hoque MM, Abdelazim H, Jenkins-Houk C, Wright D, Patel BM, Chappell JC. The cerebral microvasculature: Basic and clinical perspectives on stroke and glioma. Microcirculation 2021; 28:e12671. [PMID: 33171539 PMCID: PMC11064683 DOI: 10.1111/micc.12671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/13/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022]
Abstract
Microvascular networks are vital components of the cardiovascular system, performing many key roles in maintaining the health and homeostasis of the tissues and organs in which they develop. As discussed in this review, the molecular and cellular components within the microcirculation orchestrate critical processes to establish functional capillary beds, including organization of endothelial cell (EC) polarity, guiding investment of vascular pericytes (PCs), and building the specialized extracellular matrix (ECM) that comprises the vascular basement membrane (vBM). Herein, we further discuss the unique features of the microvasculature in the central nervous system (CNS), focusing on the cells contributing to the neurovascular unit (NVU) that form and maintain the blood-brain barrier (BBB). With a focus on vascular PCs, we offer basic and clinical perspectives on neurovascular-related pathologies that involve defects within the cerebral microvasculature. Specifically, we present microvascular anomalies associated with glioblastoma multiforme (GBM) including defects in vascular-immune cell interactions and associated clinical therapies targeting microvessels (ie, vascular-disrupting/anti-angiogenic agents and focused ultrasound). We also discuss the involvement of the microcirculation in stroke responses and potential therapeutic approaches. Our goal was to compare the cellular and molecular changes that occur in the microvasculature and NVU, and to provide a commentary on factors driving disease progression in GBM and stroke. We conclude with a forward-looking perspective on the importance of microcirculation research in developing clinical treatments for these devastating conditions.
Collapse
Affiliation(s)
- Maruf M. Hoque
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA 24061, USA
| | - Hanaa Abdelazim
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA 24061, USA
| | | | - Dawn Wright
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Biraj M. Patel
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Radiology, Carilion Clinic, Roanoke, VA, 24016, USA
| | - John C. Chappell
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
23
|
Peixoto A, Cotton S, Santos LL, Ferreira JA. The Tumour Microenvironment and Circulating Tumour Cells: A Partnership Driving Metastasis and Glycan-Based Opportunities for Cancer Control. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:1-33. [PMID: 34664231 DOI: 10.1007/978-3-030-73119-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Circulating tumour cells (CTC) are rare cells that actively detach or are shed from primary tumours into the lymph and blood. Some CTC subpopulations gain the capacity to survive, home and colonize distant locations, forming metastasis. This results from a multifactorial process in which cancer cells optimize motility, invasion, immune escape and cooperative relationships with microenvironmental cues. Here we present evidences of a self-fuelling molecular crosstalk between cancer cells and the tumour stroma supporting the main milestones leading to metastasis. We discuss how the tumour microenvironment supports pre-metastatic niches and CTC development and ultimately dictates CTC fate in targeted organs. Finally, we highlight the key role played by protein glycosylation in metastasis development, its prompt response to microenvironmental stimuli and the tremendous potential of glycan-based molecular signatures for liquid biopsies and targeted therapeutics.
Collapse
Affiliation(s)
- Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal. .,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal. .,Institute for Research and Innovation in Health (i3s), University of Porto, Porto, Portugal. .,Institute for Biomedical Engineering (INEB), Porto, Portugal. .,Porto Comprehensive Cancer Centre (P.ccc), Porto, Portugal.
| | - Sofia Cotton
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Institute for Research and Innovation in Health (i3s), University of Porto, Porto, Portugal.,Institute for Biomedical Engineering (INEB), Porto, Portugal.,Porto Comprehensive Cancer Centre (P.ccc), Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Porto Comprehensive Cancer Centre (P.ccc), Porto, Portugal.,Department of Surgical Oncology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Porto Comprehensive Cancer Centre (P.ccc), Porto, Portugal
| |
Collapse
|
24
|
Bai J, Haase K, Roberts JJ, Hoffmann J, Nguyen HT, Wan Z, Zhang S, Sarker B, Friedman N, Ristić-Lehmann Č, Kamm RD. A novel 3D vascular assay for evaluating angiogenesis across porous membranes. Biomaterials 2020; 268:120592. [PMID: 33348261 DOI: 10.1016/j.biomaterials.2020.120592] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Microfluidic technology has been extensively applied to model the functional units of human organs and tissues. Since vasculature is a key component of any functional tissue, a variety of techniques to mimic vasculature in vitro have been developed to address complex physiological and pathological processes in 3D tissues. Herein, we developed a novel, in vitro, microfluidic-based model to probe microvasculature growth into and across implanted porous membranes. Using ePTFE and polycarbonate as examples, we characterize the vascularization potential of these thin porous membranes using this device. This tool will allow for the assessment of porous materials early in their development, prior to their use for encapsulating implants or drugs, while minimizing the need for animal studies. Employing quantitative morphometric analysis and measurements of vascular permeability, we demonstrate our model to be an effective platform for evaluation of angiogenic potential of an implanted membrane biomaterial. Results show that endothelial cells can either migrate as single cells or form continuous sprouts across porous membranes, which is a material structure-dependent behavior. Our model is advantageous over conventional Transwell assays as it is amenable to quantitative assessment of vascular sprouting in 3D, and in contrast to animal models it can be employed more efficiently and with real-time assessment capabilities. This new tool could be applied either to test the suitability of a wide range of biomaterials for implantation or to screen different pro-angiogenic factors for therapeutic applications, and will advance the design of new biomaterials.
Collapse
Affiliation(s)
- Jing Bai
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kristina Haase
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Justine J Roberts
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | - Joseph Hoffmann
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | - Huu Tuan Nguyen
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhengpeng Wan
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shun Zhang
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Bapi Sarker
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | - Nathan Friedman
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | | | - Roger D Kamm
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
25
|
Watson AN, Berthiaume AA, Faino AV, McDowell KP, Bhat NR, Hartmann DA, Shih AY. Mild pericyte deficiency is associated with aberrant brain microvascular flow in aged PDGFRβ +/- mice. J Cereb Blood Flow Metab 2020; 40:2387-2400. [PMID: 31987006 PMCID: PMC7820684 DOI: 10.1177/0271678x19900543] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The receptor tyrosine kinase PDGFRβ is essential for pericyte migration to the endothelium. In mice lacking one allele of PDGFRβ (PDGFRβ+/-), previous reports have described an age-dependent loss of pericytes in the brain, leading to cerebrovascular dysfunction and subsequent neurodegeneration reminiscent of that seen in Alzheimer's disease and vascular dementia. We examined 12-20-month-old PDGFRβ+/- mice to better understand how pericyte loss affects brain microvascular structure and perfusion in vivo. We observed a mild reduction of cortical pericyte number in PDGFRβ+/- mice (27% fewer cell bodies) compared to controls, but no decrease in pericyte coverage of the endothelium. This mild degree of pericyte loss caused no discernable change in cortical microvascular density, length, basal diameter or reactivity to hypercapnia. Yet, it was associated with an increase in basal blood cell velocity, primarily in pre-capillary arterioles. Taken together, our results suggest that mild pericyte loss can lead to aberrant cerebral blood flow despite a lack of apparent effect on microvascular structure and reactivity.
Collapse
Affiliation(s)
- Ashley N Watson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Andree-Anne Berthiaume
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Anna V Faino
- Children's Core for Biomedical Statistics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Konnor P McDowell
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Narayan R Bhat
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - David A Hartmann
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Andy Y Shih
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Children's Core for Biomedical Statistics, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Blood-brain barrier integrity in the pathogenesis of Alzheimer's disease. Front Neuroendocrinol 2020; 59:100857. [PMID: 32781194 DOI: 10.1016/j.yfrne.2020.100857] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) tightly controls the molecular exchange between the brain parenchyma and blood. Accumulated evidence from transgenic animal Alzheimer's disease (AD) models and human AD patients have demonstrated that BBB dysfunction is a major player in AD pathology. In this review, we discuss the role of the BBB in maintaining brain integrity and how this is mediated by crosstalk between BBB-associated cells within the neurovascular unit (NVU). We then discuss the role of the NVU, in particular its endothelial cell, pericyte, and glial cell constituents, in AD pathogenesis. The effect of substances released by the neuroendocrine system in modulating BBB function and AD pathogenesis is also discussed. We perform a systematic review of currently available AD treatments specifically targeting pericytes and BBB glial cells. In summary, this review provides a comprehensive overview of BBB dysfunction in AD and a new perspective on the development of therapeutics for AD.
Collapse
|
27
|
Zika Virus Infection Promotes Local Inflammation, Cell Adhesion Molecule Upregulation, and Leukocyte Recruitment at the Blood-Brain Barrier. mBio 2020; 11:mBio.01183-20. [PMID: 32753493 PMCID: PMC7407083 DOI: 10.1128/mbio.01183-20] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The blood-brain barrier (BBB) largely prevents toxins and pathogens from accessing the brain. Some viruses have the ability to cross this barrier and replicate in the central nervous system (CNS). Zika virus (ZIKV) was responsible in 2015 to 2016 for a major epidemic in South America and was associated in some cases with neurological impairments. Here, we characterized some of the mechanisms behind its neuroinvasion using an innovative in vitro human BBB model. ZIKV efficiently replicated, was released on the BBB parenchyma side, and triggered subtle modulation of BBB integrity as well as an upregulation of inflammatory and cell adhesion molecules (CAMs), which in turn favored leukocyte recruitment. Finally, we showed that ZIKV-infected mouse models displayed similar CAM upregulation and that soluble CAMs were increased in plasma samples from ZIKV-infected patients. Our observations suggest a complex interplay between ZIKV and the BBB, which may trigger local inflammation, leukocyte recruitment, and possible cerebral vasculature impairment.IMPORTANCE Zika virus (ZIKV) can be associated with neurological impairment in children and adults. To reach the central nervous system, viruses have to cross the blood-brain barrier (BBB), a multicellular system allowing a tight separation between the bloodstream and the brain. Here, we show that ZIKV infects cells of the BBB and triggers a subtle change in its permeability. Moreover, ZIKV infection leads to the production of inflammatory molecules known to modulate BBB integrity and participate in immune cell attraction. The virus also led to the upregulation of cellular adhesion molecules (CAMs), which in turn favored immune cell binding to the BBB and potentially increased infiltration into the brain. These results were also observed in a mouse model of ZIKV infection. Furthermore, plasma samples from ZIKV-infected patients displayed an increase in CAMs, suggesting that this mechanism could be involved in neuroinflammation triggered by ZIKV.
Collapse
|
28
|
Jeske R, Albo J, Marzano M, Bejoy J, Li Y. Engineering Brain-Specific Pericytes from Human Pluripotent Stem Cells. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:367-382. [PMID: 32571167 PMCID: PMC7462039 DOI: 10.1089/ten.teb.2020.0091] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Pericytes (PCs) are a type of perivascular cells that surround endothelial cells of small blood vessels. In the brain, PCs show heterogeneity depending on their position within the vasculature. As a result, PC interactions with surrounding endothelial cells, astrocytes, and neuron cells play a key role in a wide array of neurovascular functions such as regulating blood-brain barrier (BBB) permeability, cerebral blood flow, and helping to facilitate the clearance of toxic cellular molecules. Therefore, a reliable method of engineering brain-specific PCs from human induced pluripotent stem cells (hiPSCs) is critical in neurodegenerative disease modeling. This review summarizes brain-specific PC differentiation of hiPSCs through mesoderm and neural crest induction. Key signaling pathways (platelet-derived growth factor-B [PDGF-B], transforming growth factor [TGF]-β, and Notch signaling) regulating PC function, PC interactions with adjacent cells, and PC differentiation from hiPSCs are also discussed. Specifically, PDGF-BB-platelet-derived growth factor receptor β signaling promotes PC cell survival, TGF-β signal transduction facilitates PC attachment to endothelial cells, and Notch signaling is critical in vascular development and arterial-venous specification. Furthermore, current challenges facing the use of hiPSC-derived PCs are discussed, and their ongoing uses in neurodegenerative disease modeling are identified. Further investigations into PCs and surrounding cell interactions are needed to characterize the roles of brain PCs in various neurodegenerative disorders. Impact statement This article summarizes the work related to brain-specific pericytes (PCs) derived from human pluripotent stem cells (hPSCs). In particular, key signaling pathways regulating PC function, PC interactions with adjacent cells, and PC differentiation from hPSCs were discussed. Furthermore, current challenges facing the use of hPSC-derived PCs were identified, and their ongoing uses in neurodegenerative disease modeling were discussed. The review highlights the important role of cell-cell interactions in blood-brain barrier (BBB) models and neurodegeneration. The summarized findings are significant for establishing pluripotent stem cell-based BBB models toward the applications in drug screening and disease modeling.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Jonathan Albo
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
29
|
Abrahamson EE, Ikonomovic MD. Brain injury-induced dysfunction of the blood brain barrier as a risk for dementia. Exp Neurol 2020; 328:113257. [PMID: 32092298 DOI: 10.1016/j.expneurol.2020.113257] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic physiological interface between brain parenchyma and cerebral vasculature. It is composed of closely interacting cells and signaling molecules that regulate movement of solutes, ions, nutrients, macromolecules, and immune cells into the brain and removal of products of normal and abnormal brain cell metabolism. Dysfunction of multiple components of the BBB occurs in aging, inflammatory diseases, traumatic brain injury (TBI, severe or mild repetitive), and in chronic degenerative dementing disorders for which aging, inflammation, and TBI are considered risk factors. BBB permeability changes after TBI result in leakage of serum proteins, influx of immune cells, perivascular inflammation, as well as impairment of efflux transporter systems and accumulation of aggregation-prone molecules involved in hallmark pathologies of neurodegenerative diseases with dementia. In addition, cerebral vascular dysfunction with persistent alterations in cerebral blood flow and neurovascular coupling contribute to brain ischemia, neuronal degeneration, and synaptic dysfunction. While the idea of TBI as a risk factor for dementia is supported by many shared pathological features, it remains a hypothesis that needs further testing in experimental models and in human studies. The current review focusses on pathological mechanisms shared between TBI and neurodegenerative disorders characterized by accumulation of pathological protein aggregates, such as Alzheimer's disease and chronic traumatic encephalopathy. We discuss critical knowledge gaps in the field that need to be explored to clarify the relationship between TBI and risk for dementia and emphasize the need for longitudinal in vivo studies using imaging and biomarkers of BBB dysfunction in people with single or multiple TBI.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
30
|
Payne LB, Zhao H, James CC, Darden J, McGuire D, Taylor S, Smyth JW, Chappell JC. The pericyte microenvironment during vascular development. Microcirculation 2019; 26:e12554. [PMID: 31066166 PMCID: PMC6834874 DOI: 10.1111/micc.12554] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/22/2022]
Abstract
Vascular pericytes provide critical contributions to the formation and integrity of the blood vessel wall within the microcirculation. Pericytes maintain vascular stability and homeostasis by promoting endothelial cell junctions and depositing extracellular matrix (ECM) components within the vascular basement membrane, among other vital functions. As their importance in sustaining microvessel health within various tissues and organs continues to emerge, so does their role in a number of pathological conditions including cancer, diabetic retinopathy, and neurological disorders. Here, we review vascular pericyte contributions to the development and remodeling of the microcirculation, with a focus on the local microenvironment during these processes. We discuss observations of their earliest involvement in vascular development and essential cues for their recruitment to the remodeling endothelium. Pericyte involvement in the angiogenic sprouting context is also considered with specific attention to crosstalk with endothelial cells such as through signaling regulation and ECM deposition. We also address specific aspects of the collective cell migration and dynamic interactions between pericytes and endothelial cells during angiogenic sprouting. Lastly, we discuss pericyte contributions to mechanisms underlying the transition from active vessel remodeling to the maturation and quiescence phase of vascular development.
Collapse
Affiliation(s)
- Laura Beth Payne
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
| | - Huaning Zhao
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061, USA
| | - Carissa C. James
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jordan Darden
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - David McGuire
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sarah Taylor
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
| | - James W. Smyth
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Department of Biological Sciences, College of Science, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061, USA
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - John C. Chappell
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061, USA
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| |
Collapse
|
31
|
Hayden MR. Type 2 Diabetes Mellitus Increases The Risk of Late-Onset Alzheimer's Disease: Ultrastructural Remodeling of the Neurovascular Unit and Diabetic Gliopathy. Brain Sci 2019; 9:brainsci9100262. [PMID: 31569571 PMCID: PMC6826500 DOI: 10.3390/brainsci9100262] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/17/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) and late-onset Alzheimer’s disease–dementia (LOAD) are increasing in global prevalence and current predictions indicate they will only increase over the coming decades. These increases may be a result of the concurrent increases of obesity and aging. T2DM is associated with cognitive impairments and metabolic factors, which increase the cellular vulnerability to develop an increased risk of age-related LOAD. This review addresses possible mechanisms due to obesity, aging, multiple intersections between T2DM and LOAD and mechanisms for the continuum of progression. Multiple ultrastructural images in female diabetic db/db models are utilized to demonstrate marked cellular remodeling changes of mural and glia cells and provide for the discussion of functional changes in T2DM. Throughout this review multiple endeavors to demonstrate how T2DM increases the vulnerability of the brain’s neurovascular unit (NVU), neuroglia and neurons are presented. Five major intersecting links are considered: i. Aging (chronic age-related diseases); ii. metabolic (hyperglycemia advanced glycation end products and its receptor (AGE/RAGE) interactions and hyperinsulinemia-insulin resistance (a linking linchpin); iii. oxidative stress (reactive oxygen–nitrogen species); iv. inflammation (peripheral macrophage and central brain microglia); v. vascular (macrovascular accelerated atherosclerosis—vascular stiffening and microvascular NVU/neuroglial remodeling) with resulting impaired cerebral blood flow.
Collapse
Affiliation(s)
- Melvin R Hayden
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO 65212, USA.
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
32
|
Osorio C, Kanukuntla T, Diaz E, Jafri N, Cummings M, Sfera A. The Post-amyloid Era in Alzheimer's Disease: Trust Your Gut Feeling. Front Aging Neurosci 2019; 11:143. [PMID: 31297054 PMCID: PMC6608545 DOI: 10.3389/fnagi.2019.00143] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022] Open
Abstract
The amyloid hypothesis, the assumption that beta-amyloid toxicity is the primary cause of neuronal and synaptic loss, has been the mainstream research concept in Alzheimer's disease for the past two decades. Currently, this model is quietly being replaced by a more holistic, “systemic disease” paradigm which, like the aging process, affects multiple body tissues and organs, including the gut microbiota. It is well-established that inflammation is a hallmark of cellular senescence; however, the infection-senescence link has been less explored. Microbiota-induced senescence is a gradually emerging concept promoted by the discovery of pathogens and their products in Alzheimer's disease brains associated with senescent neurons, glia, and endothelial cells. Infectious agents have previously been associated with Alzheimer's disease, but the cause vs. effect issue could not be resolved. A recent study may have settled this debate as it shows that gingipain, a Porphyromonas gingivalis toxin, can be detected not only in Alzheimer's disease but also in the brains of older individuals deceased prior to developing the illness. In this review, we take the position that gut and other microbes from the body periphery reach the brain by triggering intestinal and blood-brain barrier senescence and disruption. We also surmise that novel Alzheimer's disease findings, including neuronal somatic mosaicism, iron dyshomeostasis, aggressive glial phenotypes, and loss of aerobic glycolysis, can be explained by the infection-senescence model. In addition, we discuss potential cellular senescence targets and therapeutic strategies, including iron chelators, inflammasome inhibitors, senolytic antibiotics, mitophagy inducers, and epigenetic metabolic reprograming.
Collapse
Affiliation(s)
- Carolina Osorio
- Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Tulasi Kanukuntla
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Eddie Diaz
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Nyla Jafri
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Michael Cummings
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Adonis Sfera
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| |
Collapse
|
33
|
James AW, Péault B. Perivascular Mesenchymal Progenitors for Bone Regeneration. J Orthop Res 2019; 37:1221-1228. [PMID: 30908717 PMCID: PMC6546547 DOI: 10.1002/jor.24284] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal progenitor cells reside in all assayed vascularized tissues, and are broadly conceptualized to participate in homeostasis/renewal and repair. The application of mesenchymal progenitor cells has been studied for diverse orthopaedic conditions related to skeletal degeneration, regeneration, and tissue fabrication. One common niche for mesenchymal progenitors is the perivascular space, and in both mouse and human tissues, perivascular progenitor cells have been isolated and characterized. Of these "perivascular stem cells" or PSC, pericytes are the most commonly studied cells. Multiple studies have demonstrated the regenerative properties of PSC when applied to bone, including direct osteochondral differentiation, paracrine-induced osteogenesis and vasculogenesis, and immunomodulatory functions. The confluence of these effects have resulted in efficacious bone regeneration across several preclinical models. Yet, key topics of research in perivascular progenitors highlight our lack of knowledge regarding these cell populations. These ongoing areas of study include cellular diversity within the perivascular niche, tissue-specific properties of PSC, and factors that influence PSC-mediated regenerative potential. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1221-1228, 2019.
Collapse
Affiliation(s)
- Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA 90095, USA
| | - Bruno Péault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA 90095, USA,Center For Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
34
|
Silva AR, Gonçalves-de-Albuquerque CF, Pérez AR, Carvalho VDF. Immune-endocrine interactions related to a high risk of infections in chronic metabolic diseases: The role of PPAR gamma. Eur J Pharmacol 2019; 854:272-281. [PMID: 30974105 DOI: 10.1016/j.ejphar.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/11/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
Diverse disturbances in immune-endocrine circuitries are involved in the development and aggravation of several chronic metabolic diseases (CMDs), including obesity, diabetes, and metabolic syndrome. The chronic inflammatory syndrome observed in CMDs culminates in dysregulated immune responses with low microbial killing efficiency, by means low host innate immune response, and loss of ability to eliminate the pathogens, which results in a high prevalence of infectious diseases, including pneumonia, tuberculosis, and sepsis. Herein, we review evidence pointing out PPARγ as a putative player in immune-endocrine disturbances related to increased risk of infections in CMDs. Cumulated evidence indicates that PPARγ activation modulates host cells to control inflammation during CMDs because of PPARγ agonists have anti-inflammatory and pro-resolutive properties, increasing host ability to eliminate pathogen, modulating hormone production, and restoring glucose and lipid homeostasis. As such, we propose PPARγ as a putative therapeutic adjuvant for patients with CMDs to favor a better infection control.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil.
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil; Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Unirio, Brazil.
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET UNR), 2000, Rosario, Argentina.
| | - Vinicius de Frias Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
35
|
Zhao H, Chappell JC. Microvascular bioengineering: a focus on pericytes. J Biol Eng 2019; 13:26. [PMID: 30984287 PMCID: PMC6444752 DOI: 10.1186/s13036-019-0158-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/15/2019] [Indexed: 12/26/2022] Open
Abstract
Capillaries within the microcirculation are essential for oxygen delivery and nutrient/waste exchange, among other critical functions. Microvascular bioengineering approaches have sought to recapitulate many key features of these capillary networks, with an increasing appreciation for the necessity of incorporating vascular pericytes. Here, we briefly review established and more recent insights into important aspects of pericyte identification and function within the microvasculature. We then consider the importance of including vascular pericytes in various bioengineered microvessel platforms including 3D culturing and microfluidic systems. We also discuss how vascular pericytes are a vital component in the construction of computational models that simulate microcirculation phenomena including angiogenesis, microvascular biomechanics, and kinetics of exchange across the vessel wall. In reviewing these topics, we highlight the notion that incorporating pericytes into microvascular bioengineering applications will increase their utility and accelerate the translation of basic discoveries to clinical solutions for vascular-related pathologies.
Collapse
Affiliation(s)
- Huaning Zhao
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA
| | - John C Chappell
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA.,3Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016 USA
| |
Collapse
|
36
|
Barreto RSN, Romagnolli P, Cereta AD, Coimbra-Campos LMC, Birbrair A, Miglino MA. Pericytes in the Placenta: Role in Placental Development and Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:125-151. [PMID: 30937867 DOI: 10.1007/978-3-030-11093-2_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The placenta is the most variable organ, in terms of structure, among the species. Besides it, all placental types have the same function: production of viable offspring, independent of pregnancy length, litter number, or invasion level. The angiogenesis is a central mechanism for placental functionality, due to proper maternal-fetal communication and exchanges. Much is known about the vasculature structure, but little is known about vasculature development and cellular interactions. Pericytes are perivascular cells that were described to control vasculature stability and permeability. Nowadays there are several new functions discovered, such as lymphocyte modulation and activation, macrophage-like phagocytic properties, tissue regenerative and repair processes, and also the ability to modulate stem cells, majorly the hematopoietic. In parallel, placental tissues are known to be a particularly immune microenvironment and a rich stem cell niche. The pericyte function plethora could be similar in the placental microenvironment and could have a central role in placental development and homeostasis.
Collapse
Affiliation(s)
- Rodrigo S N Barreto
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Patricia Romagnolli
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Andressa Daronco Cereta
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Leda M C Coimbra-Campos
- Department of Pathology, Federal University of Minas Gerais, Pampulha, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.,Department of Pathology, Federal University of Minas Gerais, Pampulha, Belo Horizonte, Brazil
| | - Maria Angelica Miglino
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil.
| |
Collapse
|
37
|
Li W, Chen Z, Chin I, Chen Z, Dai H. The Role of VE-cadherin in Blood-brain Barrier Integrity Under Central Nervous System Pathological Conditions. Curr Neuropharmacol 2018; 16:1375-1384. [PMID: 29473514 PMCID: PMC6251046 DOI: 10.2174/1570159x16666180222164809] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/17/2017] [Accepted: 02/22/2018] [Indexed: 12/31/2022] Open
Abstract
The blood-brain barrier (BBB) is a layer between the blood circulation and neural tissue. It plays a pivotal role in maintaining the vulnerable extracellular microenvironment in the neuronal parenchyma. Neuroinflammatory events can result in BBB dysregulation by disturbing adherens junctions (AJs) and tight junctions (TJs). VE-cadherin, as one of the most im-portant components of the vascular system, is specifically responsible for the assembly of AJs and BBB architecture. Here, we present a review, which highlights recently available insights into the relationship between the neuroinflammation and BBB dysregulation. We then explore the specific interaction between VE-cadherin and BBB. Fi-nally, we discuss the changes of VE-cadherin with different neurological diseases from both experimental and clinical stud-ies. An understanding of VE-cadherin in BBB regulation may indicate that VE-cadherin can partially be a biomarker of neu-roinflammation disease and lead to novel approaches for abating BBB dysregulation under pathological conditions and the opening of the BBB following central nervous system (CNS) drug delivery.
Collapse
Affiliation(s)
- Wenlu Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China.,Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zhigang Chen
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Ian Chin
- Metcalf Science Center, Boston University, Boston, MA 02215, United States
| | - Zhong Chen
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Haibin Dai
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| |
Collapse
|
38
|
Hou Z, Wang X, Cai J, Zhang J, Hassan A, Auer M, Shi X. Platelet-Derived Growth Factor Subunit B Signaling Promotes Pericyte Migration in Response to Loud Sound in the Cochlear Stria Vascularis. J Assoc Res Otolaryngol 2018; 19:363-379. [PMID: 29869048 PMCID: PMC6081892 DOI: 10.1007/s10162-018-0670-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/19/2018] [Indexed: 12/20/2022] Open
Abstract
Normal blood supply to the cochlea is critical for hearing. Noise damages auditory sensory cells and has a marked effect on the microvasculature in the cochlear lateral wall. Pericytes in the stria vascularis (strial pericytes) are particularly vulnerable and sensitive to acoustic trauma. Exposure of NG2DsRedBAC transgenic mice (6-8 weeks old) to wide-band noise at a level of 120 dB for 3 h per day for 2 consecutive days produced a significant hearing threshold shift and caused pericytes to protrude and migrate from their normal endothelial attachment sites. The pericyte migration was associated with increased expression of platelet-derived growth factor beta (PDGF-BB). Blockade of PDGF-BB signaling with either imatinib, a potent PDGF-BB receptor (PDGFR) inhibitor, or APB5, a specific PDGFRβ blocker, significantly attenuated the pericyte migration from strial vessel walls. The PDGF-BB-mediated strial pericyte migration was further confirmed in an in vitro cell migration assay, as well as in an in vivo live animal model used in conjunction with confocal fluorescence microscopy. Pericyte migration took one of two different forms, here denoted protrusion and detachment. The protrusion is characterized by pericytes with a prominent triangular shape, or pericytes extending fine strands to neighboring capillaries. The detachment is characterized by pericyte detachment and movement away from vessels. We also found the sites of pericyte migration highly associated with regions of vascular leakage. In particular, under transmission electron microscopy (TEM), multiple vesicles at the sites of endothelial cells with loosely attached pericytes were observed. These data show that cochlear pericytes are markedly affected by acoustic trauma, causing them to display abnormal morphology. The effect of loud sound on pericytes is mediated by upregulation of PDGF-BB. Normal functioning pericytes are required for vascular stability.
Collapse
Affiliation(s)
- Zhiqiang Hou
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Xiaohan Wang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jing Cai
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jinhui Zhang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Ahmed Hassan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Manfred Auer
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
39
|
Zhao X, Eyo UB, Murguan M, Wu LJ. Microglial interactions with the neurovascular system in physiology and pathology. Dev Neurobiol 2018; 78:604-617. [PMID: 29318762 PMCID: PMC5980686 DOI: 10.1002/dneu.22576] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/01/2018] [Accepted: 01/06/2018] [Indexed: 01/11/2023]
Abstract
Microglia as immune cells of the central nervous system (CNS) play significant roles not only in pathology but also in physiology, such as shaping of the CNS during development and its proper maintenance in maturity. Emerging research is showing a close association between microglia and the neurovasculature that is critical for brain energy supply. In this review, we summarize the current literature on microglial interaction with the vascular system in the normal and diseased brain. First, we highlight data that indicate interesting potential involvement of microglia in developmental angiogenesis. Then we discuss the evidence for microglial participation with the vasculature in neuropathologies from brain tumors to acute injuries such as ischemic stroke to chronic neurodegenerative conditions. We conclude by suggesting future areas of research to advance the field in light of current technical progress and outstanding questions. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 604-617, 2018.
Collapse
Affiliation(s)
- Xiaoliang Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
| | - Ukpong B. Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Madhuvika Murguan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
40
|
Meyers CA, Xu J, Zhang L, Asatrian G, Ding C, Yan N, Broderick K, Sacks J, Goyal R, Zhang X, Ting K, Péault B, Soo C, James AW. Early Immunomodulatory Effects of Implanted Human Perivascular Stromal Cells During Bone Formation. Tissue Eng Part A 2018; 24:448-457. [PMID: 28683667 PMCID: PMC5833257 DOI: 10.1089/ten.tea.2017.0023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/06/2017] [Indexed: 01/20/2023] Open
Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesodermal progenitor cell population defined by their perivascular residence. PSC are most commonly derived from subcutaneous adipose tissue, and recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting-derived cell population for bone tissue engineering. Specifically, purified PSC induce greater bone formation than unpurified stroma taken from the same patient sample. In this study, we examined the differences in early innate immune response to human PSC or unpurified stroma (stromal vascular fraction [SVF]) during the in vivo process of bone formation. Briefly, SVF or PSC from the same patient sample were implanted intramuscularly in the hindlimb of severe combined immunodeficient (SCID) mice using an osteoinductive demineralized bone matrix carrier. Histological examination of early inflammatory infiltrates was examined by hematoxylin and eosin and immunohistochemical staining (Ly-6G, F4/80). Results showed significantly greater neutrophilic and macrophage infiltrates within and around SVF in comparison to PSC-laden implants. Differences in early postoperative inflammation among SVF-laden implants were associated with reduced osteogenic differentiation and bone formation. Similar findings were recapitulated with PSC implantation in immunocompetent mice. Exaggerated postoperative inflammation was associated with increased IL-1α, IL-1β, IFN-γ, and TNF-α gene expression among SVF samples, and conversely increased IL-6 and IL-10 expression among PSC samples. These data document a robust immunomodulatory effect of implanted PSC, and an inverse correlation between host inflammatory cell infiltration and stromal progenitor cell-mediated ossification.
Collapse
Affiliation(s)
- Carolyn A. Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Lei Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, PR China
| | - Greg Asatrian
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Catherine Ding
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Noah Yan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Kristen Broderick
- Department of Plastic Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Justin Sacks
- Department of Plastic Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Raghav Goyal
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Bruno Péault
- Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- UCLA Orthopedic Hospital Department of Orthopedic Surgery and the Orthopedic Hospital Research Center, Los Angeles, California
| | - Chia Soo
- UCLA Orthopedic Hospital Department of Orthopedic Surgery and the Orthopedic Hospital Research Center, Los Angeles, California
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
- UCLA Orthopedic Hospital Department of Orthopedic Surgery and the Orthopedic Hospital Research Center, Los Angeles, California
| |
Collapse
|
41
|
Giannoni P, Badaut J, Dargazanli C, Fayd'Herbe De Maudave A, Klement W, Costalat V, Marchi N. The pericyte-glia interface at the blood-brain barrier. Clin Sci (Lond) 2018; 132:361-374. [PMID: 29439117 DOI: 10.1042/cs20171634] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
Abstract
The cerebrovasculature is a multicellular structure with varying rheological and permeability properties. The outer wall of the brain capillary endothelium is enclosed by pericytes and astrocyte end feet, anatomically assembled to guarantee barrier functions. We, here, focus on the pericyte modifications occurring in disease conditions, reviewing evidence supporting the interplay amongst pericytes, the endothelium, and glial cells in health and pathology. Deconstruction and reactivity of pericytes and glial cells around the capillary endothelium occur in response to traumatic brain injury, epilepsy, and neurodegenerative disorders, impacting vascular permeability and participating in neuroinflammation. As this represents a growing field of research, addressing the multicellular reorganization occurring at the outer wall of the blood-brain barrier (BBB) in response to an acute insult or a chronic disease could disclose novel disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
| | - Jerome Badaut
- Laboratory of Brain Molecular Imaging, CNRS UMR5287, University of Bordeaux, France
- Basic Science Departments, Loma Linda University School of Medicine, CA, U.S.A
| | - Cyril Dargazanli
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Alexis Fayd'Herbe De Maudave
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Wendy Klement
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Vincent Costalat
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
42
|
Jackson S, ElAli A, Virgintino D, Gilbert MR. Blood-brain barrier pericyte importance in malignant gliomas: what we can learn from stroke and Alzheimer's disease. Neuro Oncol 2017; 19:1173-1182. [PMID: 28541444 PMCID: PMC5570196 DOI: 10.1093/neuonc/nox058] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pericyte, a constitutive component of the central nervous system, is a poorly understood cell type that envelops the endothelial cell with the intended purpose of regulating vascular flow and endothelial cell permeability. Previous studies of pericyte function have been limited to a small number of disease processes such as ischemic stroke and Alzheimer's disease. Recently, publications have postulated a link between glioma stem cell differentiation and pericyte function. These studies suggest that there may be an important interaction of pericytes with tumor cells and other components of the tumor microenvironment in malignant primary glial neoplasms, most notably glioblastoma. This potential cellular interaction underscores the need to pursue more investigations of pericytes in malignant brain tumor biology. In this review, we summarize the functional roles of pericytes, particularly focusing on changes in pericyte biology during response to immune cells, inflammation, and hypoxic conditions. The information presented is based on the available data from studies of pericyte function in other central nervous system diseases but will serve as a foundation for research investigations to further understand the role of pericytes in malignant gliomas.
Collapse
Affiliation(s)
- Sadhana Jackson
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Ayman ElAli
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Mark R Gilbert
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
43
|
Novel Regenerative Therapies Based on Regionally Induced Multipotent Stem Cells in Post-Stroke Brains: Their Origin, Characterization, and Perspective. Transl Stroke Res 2017; 8:515-528. [PMID: 28744717 DOI: 10.1007/s12975-017-0556-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/07/2017] [Accepted: 07/18/2017] [Indexed: 12/16/2022]
Abstract
Brain injuries such as ischemic stroke cause severe neural loss. Until recently, it was believed that post-ischemic areas mainly contain necrotic tissue and inflammatory cells. However, using a mouse model of cerebral infarction, we demonstrated that stem cells develop within ischemic areas. Ischemia-induced stem cells can function as neural progenitors; thus, we initially named them injury/ischemia-induced neural stem/progenitor cells (iNSPCs). However, because they differentiate into more than neural lineages, we now refer to them as ischemia-induced multipotent stem cells (iSCs). Very recently, we showed that putative iNSPCs/iSCs are present within post-stroke areas in human brains. Because iNSPCs/iSCs isolated from mouse and human ischemic tissues can differentiate into neuronal lineages in vitro, it is possible that a clearer understanding of iNSPC/iSC profiles and the molecules that regulate iNSPC/iSC fate (e.g., proliferation, differentiation, and survival) would make it possible to perform neural regeneration/repair in patients following stroke. In this article, we introduce the origin and traits of iNSPCs/iSCs based on our reports and recent viewpoints. We also discuss their possible contribution to neurogenesis through endogenous and exogenous iNSPC/iSC therapies following ischemic stroke.
Collapse
|
44
|
Zhang Y, Zhang Y, Bai Y, Chao J, Hu G, Chen X, Yao H. Involvement of PUMA in pericyte migration induced by methamphetamine. Exp Cell Res 2017; 356:28-39. [PMID: 28408317 DOI: 10.1016/j.yexcr.2017.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 12/21/2022]
Abstract
Mounting evidence indicates that methamphetamine causes blood-brain barrier damage, with emphasis on endothelial cells. The role of pericytes in methamphetamine-induced BBB damage remains unknown. Our study demonstrated that methamphetamine increased the migration of pericytes from the endothelial basement membrane. However, the detailed mechanisms underlying this process remain poorly understood. Thus, we examined the molecular mechanisms involved in methamphetamine-induced pericyte migration. The results showed that exposure of C3H/10T1/2 cells and HBVPs to methamphetamine increased PUMA expression via activation of the sigma-1 receptor, MAPK and Akt/PI3K pathways. Moreover, methamphetamine treatment resulted in the increased migration of C3H/10T1/2 cells and HBVPs. Knockdown of PUMA in pericytes transduced with PUMA siRNA attenuated the methamphetamine-induced increase in cell migration through attenuation of integrin and tyrosine kinase mechanisms, implicating a role of PUMA in the migration of C3H/10T1/2 cells and HBVPs. This study has demonstrated that methamphetamine-mediated pericytes migration involves PUMA up-regulation. Thus, targeted studies of PUMA could provide insights to facilitate the development of a potential therapeutic approach for alleviation of methamphetamine-induced pericyte migration.
Collapse
Affiliation(s)
- Yanhong Zhang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Yuan Zhang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Ying Bai
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Jie Chao
- Department of Physiology, Medical School of Southeast University, Nanjing, China.
| | - Gang Hu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China.
| | - Xufeng Chen
- Department of Emergency, Jiangsu Province Hospital, Nanjing, Jiangsu, China.
| | - Honghong Yao
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
45
|
Abstract
While stroke research represents the primary interface between circulation and brain research, the hemostasis system also carries a pivotal role in the mechanism of vascular brain injury. The complex interrelated events triggered by the energy crisis have a specific spatial and temporal pattern arching from the initial damage to the final events of brain repair. The complexity of the pathophysiology make it difficult to model this disease, therefore it is challenging to find appropriate therapeutic targets. The ever-persistent antagonism between the positive results of drug candidates in the experimental stroke models and the failures of the clinical trials prompts changes in the research strategy, especially in the field of potential neuroprotective therapies. System biology approach could initiate new directions in the future for both preclinical and clinical research. Incentive methods aimed at anti-apoptosis mechanisms and the augmentation of post-ischemic brain repair could benefit the facts, that these processes can be targeted much longer following the cell-necrosis in the hyper-acute phase. Sequential monitoring of candidate genes and proteins responsible for stroke progression and post-stroke repair seems to be useful both in therapeutic target-identification, and in clinical testing. Understanding the mechanism behind the effect of selegiline and other drugs capable of activating the anti-apoptotic gene expression could help to find new approaches to enhance the regenerative potential in the remodeling of neuronal and microvascular networks.
Collapse
Affiliation(s)
- Z Nagy
- Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Városmajor Street 68, 1122, Hungary; National Institute of Clinical Neurosciences, Budapest, Amerikai Street 57, 1145, Hungary.
| | - S Nardai
- Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Városmajor Street 68, 1122, Hungary; National Institute of Clinical Neurosciences, Budapest, Amerikai Street 57, 1145, Hungary
| |
Collapse
|
46
|
Shi X, Zhang W, Yin L, Chilian WM, Krieger J, Zhang P. Vascular precursor cells in tissue injury repair. Transl Res 2017; 184:77-100. [PMID: 28284670 PMCID: PMC5429880 DOI: 10.1016/j.trsl.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
Abstract
Vascular precursor cells include stem cells and progenitor cells giving rise to all mature cell types in the wall of blood vessels. When tissue injury occurs, local hypoxia and inflammation result in the generation of vasculogenic mediators which orchestrate migration of vascular precursor cells from their niche environment to the site of tissue injury. The intricate crosstalk among signaling pathways coordinates vascular precursor cell proliferation and differentiation during neovascularization. Establishment of normal blood perfusion plays an essential role in the effective repair of the injured tissue. In recent years, studies on molecular mechanisms underlying the regulation of vascular precursor cell function have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches to treat chronic wounds and ischemic diseases in vital organ systems. Verification of safety and establishment of specific guidelines for the clinical application of vascular precursor cell-based therapy remain major challenges in the field.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Weihong Zhang
- Department of Basic Medicine, School of Nursing, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Liya Yin
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Jessica Krieger
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
47
|
Lauschke K, Frederiksen L, Hall VJ. Paving the Way Toward Complex Blood-Brain Barrier Models Using Pluripotent Stem Cells. Stem Cells Dev 2017; 26:857-874. [PMID: 28398169 DOI: 10.1089/scd.2017.0003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A tissue with great need to be modeled in vitro is the blood-brain barrier (BBB). The BBB is a tight barrier that covers all blood vessels in the brain and separates the brain microenvironment from the blood system. It consists of three cell types [neurovascular unit (NVU)] that contribute to the unique tightness and selective permeability of the BBB and has been shown to be disrupted in many diseases and brain disorders, such as vascular dementia, stroke, multiple sclerosis, and Alzheimer's disease. Given the progress that pluripotent stem cells (PSCs) have made in the past two decades, it is now possible to produce many cell types from the BBB and even partially recapitulate this complex tissue in vitro. In this review, we summarize the most recent developments in PSC differentiation and modeling of the BBB. We also suggest how patient-specific human-induced PSCs could be used to model BBB dysfunction in the future. Lastly, we provide perspectives on how to improve production of the BBB in vitro, for example by improving pericyte differentiation protocols and by better modeling the NVU in the dish.
Collapse
Affiliation(s)
- Karin Lauschke
- 1 National Food Institute, Technical University of Denmark , Kongens Lyngby, Denmark
- 2 Department of Micro- and Nanotechnology, Technical University of Denmark , Kongens Lyngby, Denmark
| | - Lise Frederiksen
- 3 Faculty of Health and Medical Sciences, University of Copenhagen , København N, Denmark
| | - Vanessa Jane Hall
- 4 Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Frederiksberg C, Denmark
| |
Collapse
|
48
|
Pearn ML, Niesman IR, Egawa J, Sawada A, Almenar-Queralt A, Shah SB, Duckworth JL, Head BP. Pathophysiology Associated with Traumatic Brain Injury: Current Treatments and Potential Novel Therapeutics. Cell Mol Neurobiol 2017; 37:571-585. [PMID: 27383839 DOI: 10.1007/s10571-016-0400-1] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death of young people in the developed world. In the United States alone, 1.7 million traumatic events occur annually accounting for 50,000 deaths. The etiology of TBI includes traffic accidents, falls, gunshot wounds, sports, and combat-related events. TBI severity ranges from mild to severe. TBI can induce subtle changes in molecular signaling, alterations in cellular structure and function, and/or primary tissue injury, such as contusion, hemorrhage, and diffuse axonal injury. TBI results in blood-brain barrier (BBB) damage and leakage, which allows for increased extravasation of immune cells (i.e., increased neuroinflammation). BBB dysfunction and impaired homeostasis contribute to secondary injury that occurs from hours to days to months after the initial trauma. This delayed nature of the secondary injury suggests a potential therapeutic window. The focus of this article is on the (1) pathophysiology of TBI and (2) potential therapies that include biologics (stem cells, gene therapy, peptides), pharmacological (anti-inflammatory, antiepileptic, progrowth), and noninvasive (exercise, transcranial magnetic stimulation). In final, the review briefly discusses membrane/lipid rafts (MLR) and the MLR-associated protein caveolin (Cav). Interventions that increase Cav-1, MLR formation, and MLR recruitment of growth-promoting signaling components may augment the efficacy of pharmacologic agents or already existing endogenous neurotransmitters and neurotrophins that converge upon progrowth signaling cascades resulting in improved neuronal function after injury.
Collapse
Affiliation(s)
- Matthew L Pearn
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Ingrid R Niesman
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Junji Egawa
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Atsushi Sawada
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Sameer B Shah
- UCSD Departments of Orthopaedic Surgery and Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Josh L Duckworth
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Brian P Head
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA.
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
49
|
Hu X, De Silva TM, Chen J, Faraci FM. Cerebral Vascular Disease and Neurovascular Injury in Ischemic Stroke. Circ Res 2017; 120:449-471. [PMID: 28154097 PMCID: PMC5313039 DOI: 10.1161/circresaha.116.308427] [Citation(s) in RCA: 280] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022]
Abstract
The consequences of cerebrovascular disease are among the leading health issues worldwide. Large and small cerebral vessel disease can trigger stroke and contribute to the vascular component of other forms of neurological dysfunction and degeneration. Both forms of vascular disease are driven by diverse risk factors, with hypertension as the leading contributor. Despite the importance of neurovascular disease and subsequent injury after ischemic events, fundamental knowledge in these areas lag behind our current understanding of neuroprotection and vascular biology in general. The goal of this review is to address select key structural and functional changes in the vasculature that promote hypoperfusion and ischemia, while also affecting the extent of injury and effectiveness of therapy. In addition, as damage to the blood-brain barrier is one of the major consequences of ischemia, we discuss cellular and molecular mechanisms underlying ischemia-induced changes in blood-brain barrier integrity and function, including alterations in endothelial cells and the contribution of pericytes, immune cells, and matrix metalloproteinases. Identification of cell types, pathways, and molecules that control vascular changes before and after ischemia may result in novel approaches to slow the progression of cerebrovascular disease and lessen both the frequency and impact of ischemic events.
Collapse
Affiliation(s)
- Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - T. Michael De Silva
- Biomedicine Discovery Institute, Department of Pharmacology, 9 Ancora Imparo Way, Monash University, Clayton, Vic, Australia
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Frank M. Faraci
- Departments of Internal Medicine and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City Veterans Affairs Healthcare System, Iowa City, IA, USA
| |
Collapse
|
50
|
Nishikawa K, Furube E, Morita S, Horii-Hayashi N, Nishi M, Miyata S. Structural Reconstruction of the Perivascular Space in the Adult Mouse Neurohypophysis During an Osmotic Stimulation. J Neuroendocrinol 2017; 29. [PMID: 28072496 DOI: 10.1111/jne.12456] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/19/2016] [Accepted: 01/08/2017] [Indexed: 12/11/2022]
Abstract
Oxytocin (OXT) and arginine vasopressin (AVP) neuropeptides in the neurohypophysis (NH) control lactation and body fluid homeostasis, respectively. Hypothalamic neurosecretory neurones project their axons from the supraoptic and paraventricular nuclei to the NH to make contact with the vascular surface and release OXT and AVP. The neurohypophysial vascular structure is unique because it has a wide perivascular space between the inner and outer basement membranes. However, the significance of this unique vascular structure remains unclear; therefore, we aimed to determine the functional significance of the perivascular space and its activity-dependent changes during salt loading in adult mice. The results obtained revealed that pericytes were the main resident cells and defined the profile of the perivascular space. Moreover, pericytes sometimes extended their cellular processes or 'perivascular protrusions' into neurohypophysial parenchyma between axonal terminals. The vascular permeability of low-molecular-weight (LMW) molecules was higher at perivascular protrusions than at the smooth vascular surface. Axonal terminals containing OXT and AVP were more likely to localise at perivascular protrusions than at the smooth vascular surface. Chronic salt loading with 2% NaCl significantly induced prominent changes in the shape of pericytes and also increased the number of perivascular protrusions and the surface area of the perivascular space together with elevations in the vascular permeability of LMW molecules. Collectively, these results indicate that the perivascular space of the NH acts as the main diffusion route for OXT and AVP and, in addition, changes in the shape of pericytes and perivascular reconstruction occur in response to an increased demand for neuropeptide release.
Collapse
Affiliation(s)
- K Nishikawa
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - E Furube
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - S Morita
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
- Department of Anatomy and Neuroscience, Nara Medical University, Kashihara, Nara, Japan
| | - N Horii-Hayashi
- Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - M Nishi
- Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - S Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
- The Center for Advanced Insect Research Promotion (CAIRP), Kyoto Institute of Technology, Kyoto, Japan
| |
Collapse
|