1
|
Dong W, Sheng J, Cui JZM, Zhao H, Wong STC. Systems immunology insights into brain metastasis. Trends Immunol 2024; 45:903-916. [PMID: 39443266 DOI: 10.1016/j.it.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Brain metastasis poses formidable clinical challenges due to its intricate interactions with the brain's unique immune environment, often resulting in poor prognoses. This review delves into systems immunology's role in uncovering the dynamic interplay between metastatic cancer cells and brain immunity. Leveraging spatial and single-cell technologies, along with advanced computational modeling, systems immunology offers unprecedented insights into mechanisms of immune evasion and tumor proliferation. Recent studies highlight potential immunotherapeutic targets, suggesting strategies to boost antitumor immunity and counteract cancer cell evasion in the brain. Despite substantial progress, challenges persist, particularly in accurately simulating human conditions. This review underscores the need for interdisciplinary collaboration to harness systems immunology's full potential, aiming to dramatically improve outcomes for patients with brain metastasis.
Collapse
Affiliation(s)
- Wenjuan Dong
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Jianting Sheng
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Johnny Z M Cui
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Hong Zhao
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA.
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Brandon KD, Frank WE, Stroka KM. Junctions at the crossroads: the impact of mechanical cues on endothelial cell-cell junction conformations and vascular permeability. Am J Physiol Cell Physiol 2024; 327:C1073-C1086. [PMID: 39129490 PMCID: PMC11481987 DOI: 10.1152/ajpcell.00605.2023] [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: 11/08/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Cells depend on precisely regulating barrier function within the vasculature to maintain physiological stability and facilitate essential substance transport. Endothelial cells achieve this through specialized adherens and tight junction protein complexes, which govern paracellular permeability across vascular beds. Adherens junctions, anchored by vascular endothelial (VE)-cadherin and associated catenins to the actin cytoskeleton, mediate homophilic adhesion crucial for barrier integrity. In contrast, tight junctions composed of occludin, claudin, and junctional adhesion molecule A interact with Zonula Occludens proteins, reinforcing intercellular connections essential for barrier selectivity. Endothelial cell-cell junctions exhibit dynamic conformations during development, maturation, and remodeling, regulated by local biochemical and mechanical cues. These structural adaptations play pivotal roles in disease contexts such as chronic inflammation, where junctional remodeling contributes to increased vascular permeability observed in conditions from cancer to cardiovascular diseases. Conversely, the brain microvasculature's specialized junctional arrangements pose challenges for therapeutic drug delivery due to their unique molecular compositions and tight organization. This commentary explores the molecular mechanisms underlying endothelial cell-cell junction conformations and their implications for vascular permeability. By highlighting recent advances in quantifying junctional changes and understanding mechanotransduction pathways, we elucidate how physical forces from cellular contacts and hemodynamic flow influence junctional dynamics.
Collapse
Affiliation(s)
- Ken D Brandon
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
| | - William E Frank
- Department of Biology, University of Puerto Rico in Ponce, Ponce, Puerto Rico
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, United States
- Biophysics Program, University of Maryland, College Park, Maryland, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, Maryland, United States
| |
Collapse
|
3
|
Oliveira-Paula GH, Martins AC, Ferrer B, Tinkov AA, Skalny AV, Aschner M. The impact of manganese on vascular endothelium. Toxicol Res 2024; 40:501-517. [PMID: 39345740 PMCID: PMC11436708 DOI: 10.1007/s43188-024-00260-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024] Open
Abstract
Manganese (Mn) is an essential trace element involved in various physiological processes, but excessive exposure may lead to toxicity. The vascular endothelium, a monolayer of endothelial cells within blood vessels, is a primary target of Mn toxicity. This review provides a comprehensive overview of the impact of Mn on vascular endothelium, focusing on both peripheral and brain endothelial cells. In vitro studies have demonstrated that high concentrations of Mn can induce endothelial cell cytotoxicity, increase permeability, and disrupt cell-cell junctions through mechanisms involving oxidative stress, mitochondrial damage, and activation of signaling pathways, such as Smad2/3-Snail. Conversely, low concentrations of Mn may protect endothelial cells from the deleterious effects of high glucose and advanced glycation end-products. In the central nervous system, Mn can cross the blood-brain barrier (BBB) and accumulate in the brain parenchyma, leading to neurotoxicity. Several transport mechanisms, including ZIP8, ZIP14, and SPCA1, have been identified for Mn uptake by brain endothelial cells. Mn exposure can impair BBB integrity by disrupting tight junctions and increasing permeability. In vivo studies have corroborated these findings, highlighting the importance of endothelial barriers in mediating Mn toxicity in the brain and kidneys. Maintaining optimal Mn homeostasis is crucial for preserving endothelial function, and further research is needed to develop targeted therapeutic strategies to prevent or mitigate the adverse effects of Mn overexposure. Graphical Abstract
Collapse
Affiliation(s)
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003 Russia
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435 Russia
| | - Anatoly V. Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003 Russia
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435 Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| |
Collapse
|
4
|
Sugimura K, Otani T. Vertex remodeling during epithelial morphogenesis. Curr Opin Cell Biol 2024; 91:102427. [PMID: 39332144 DOI: 10.1016/j.ceb.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024]
Abstract
Epithelial cells adhere to each other via intercellular junctions that can be classified into bicellular junctions and tricellular contacts (vertices). Epithelial morphogenesis involves cell rearrangement and requires remodeling of bicellular junctions and vertices. Although our understanding of how bicellular junction mechanics drive epithelial morphogenesis has advanced, the mechanisms underlying vertex remodeling during this process have only received attention recently. In this review, we outline recent progress in our understanding of how cells reorganize cell adhesion and the cytoskeleton to trigger the displacement and resolution of cell vertices. We will also discuss how cells achieve the optimal balance between the structural flexibility and stability of their vertices. Finally, we introduce new modeling frameworks designed to analyze mechanics at cell vertices. Integration of live imaging and modeling techniques is providing new insights into the active roles of cell vertices during epithelial morphogenesis.
Collapse
Affiliation(s)
- Kaoru Sugimura
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan; Universal Biology Institute, The University of Tokyo, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan.
| | - Tetsuhisa Otani
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan; Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology (PRESTO), Saitama, Japan.
| |
Collapse
|
5
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2024:revneuro-2024-0090. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [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: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
6
|
Sun Z, Zhang B, Zhou J, Luo Y, Zhu X, Wang Y, He Y, Zheng P, Zhang L, Yang J, Wang G. Integrated Single-Cell RNA-seq and ATAC-seq Reveals Heterogeneous Differentiation of CD4 + Naive T Cell Subsets is Associated with Response to Antidepressant Treatment in Major Depressive Disorder. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308393. [PMID: 38867657 PMCID: PMC11321657 DOI: 10.1002/advs.202308393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/08/2024] [Indexed: 06/14/2024]
Abstract
The mechanism involved in major depressive disorder (MDD) is well-studied but the mechanistic origin of the heterogeneous antidepressant effect remains largely unknown. Single-cell RNA-sequencing (scRNA-seq) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) on peripheral blood mononuclear cells from 8 healthy individuals and 8 MDD patients before or after 12 weeks of antidepressant treatment is performed. scRNA-seq analysis reveals a lower proportion of naive T cells, particularly CD4+ naive T cells, in MDD patients compared to controls, and in nonresponders versus responders at the baseline. Flow cytometry data analysis of an independent cohort of 35 patients and 40 healthy individuals confirms the findings. Enrichment analysis of differentially expressed genes indicated obvious immune activation in responders. A specific activated CD4+ naive T population in responders characterized by enhanced mitogen-activated protein kinases (MAPK) pathway is identified. E-twenty six (ETS) is proposed as an upstream regulator of the MAPK pathway and heterogeneous differentiation in activated CD4+ naive T population is associated with the response to antidepressant treatment in MDD patients. A distinct immune feature manifested by CD4+ naive T cells during antidepressant treatment in MDD is identified. Collectively, this proposes the molecular mechanism that underlies the heterogeneous antidepressant outcomes for MDD.
Collapse
Affiliation(s)
- Zuoli Sun
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Bowen Zhang
- College of Life SciencesBeijing Normal UniversityBeijing100875China
| | - Jingjing Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Yanting Luo
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Xuequan Zhu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Yaping Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Yi He
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Peng Zheng
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional DiseasesThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Ling Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100069China
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental DisordersBeijing Anding HospitalCapital Medical UniversityBeijing100088China
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100069China
| |
Collapse
|
7
|
Berve K, Michel J, Tietz S, Blatti C, Ivan D, Enzmann G, Lyck R, Deutsch U, Locatelli G, Engelhardt B. Junctional adhesion molecule-A deficient mice are protected from severe experimental autoimmune encephalomyelitis. Eur J Immunol 2024; 54:e2350761. [PMID: 38566526 DOI: 10.1002/eji.202350761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.
Collapse
Affiliation(s)
- Kristina Berve
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Julia Michel
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Claudia Blatti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Daniela Ivan
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
8
|
Voges L, Weiß F, Branco AT, Fromm M, Krug SM. Expression and Localization Profiles of Tight Junction Proteins in Immune Cells Depend on Their Activation Status. Int J Mol Sci 2024; 25:4861. [PMID: 38732086 PMCID: PMC11084252 DOI: 10.3390/ijms25094861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
The ability of the immune system to combat pathogens relies on processes like antigen sampling by dendritic cells and macrophages migrating through endo- and epithelia or penetrating them with their dendrites. In addition, other immune cell subtypes also migrate through the epithelium after activation. For paracellular migration, interactions with tight junctions (TJs) are necessary, and previous studies reported TJ protein expression in several immune cells. Our investigation aimed to characterize, in more detail, the expression profiles of TJ proteins in different immune cells in both naïve and activated states. The mRNA expression analysis revealed distinct expression patterns for TJ proteins, with notable changes, mainly increases, upon activation. At the protein level, LSR appeared predominant, being constitutively present in naïve cell membranes, suggesting roles as a crucial interaction partner. Binding experiments suggested the presence of claudins in the membrane only after stimulation, and claudin-8 translocation to the membrane occurred after stimulation. Our findings suggest a dynamic TJ protein expression in immune cells, implicating diverse functions in response to stimulation, like interaction with TJ proteins or regulatory roles. While further analysis is needed to elucidate the precise roles of TJ proteins, our findings indicate important non-canonical functions of TJ proteins in immune response.
Collapse
Affiliation(s)
- Lena Voges
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Franziska Weiß
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Ana-Teresa Branco
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Michael Fromm
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Susanne M. Krug
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| |
Collapse
|
9
|
Zapata-Acevedo JF, Mantilla-Galindo A, Vargas-Sánchez K, González-Reyes RE. Blood-brain barrier biomarkers. Adv Clin Chem 2024; 121:1-88. [PMID: 38797540 DOI: 10.1016/bs.acc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.
Collapse
Affiliation(s)
- Juan F Zapata-Acevedo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Mantilla-Galindo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
10
|
Kallal N, Hugues S, Garnier L. Regulation of autoimmune-mediated neuroinflammation by endothelial cells. Eur J Immunol 2024; 54:e2350482. [PMID: 38335316 DOI: 10.1002/eji.202350482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
The CNS has traditionally been considered an immune-privileged organ, but recent studies have identified a plethora of immune cells in the choroid plexus, meninges, perivascular spaces, and cribriform plate. Although those immune cells are crucial for the maintenance of CNS homeostasis and for neural protection against infections, they can lead to neuroinflammation in some circumstances. The blood and the lymphatic vasculatures exhibit distinct structural and molecular features depending on their location in the CNS, greatly influencing the compartmentalization and the nature of CNS immune responses. In this review, we discuss how endothelial cells regulate the migration and the functions of T cells in the CNS both at steady-state and in murine models of neuroinflammation, with a special focus on the anatomical, cellular, and molecular mechanisms implicated in EAE.
Collapse
Affiliation(s)
- Neil Kallal
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Laure Garnier
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
11
|
Ruiz-Fernández I, Sánchez-Díaz R, Ortega-Sollero E, Martín P. Update on the role of T cells in cognitive impairment. Br J Pharmacol 2024; 181:799-815. [PMID: 37559406 DOI: 10.1111/bph.16214] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/03/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
The central nervous system (CNS) has long been considered an immune-privileged site, with minimal interaction between immune cells, particularly of the adaptive immune system. Previously, the presence of immune cells in this organ was primarily linked to events involving disruption of the blood-brain barrier (BBB) or inflammation. However, current research has shown that immune cells are found patrolling CNS under homeostatic conditions. Specifically, T cells of the adaptive immune system are able to cross the BBB and are associated with ageing and cognitive impairment. In addition, T-cell infiltration has been observed in pathological conditions, where inflammation correlates with poor prognosis. Despite ongoing research, the role of this population in the ageing brain under both physiological and pathological conditions is not yet fully understood. In this review, we provide an overview of the interactions between T cells and other immune and CNS parenchymal cells, and examine the molecular mechanisms by which these interactions may contribute to normal brain function and the scenarios in which disruption of these connections lead to cognitive impairment. A comprehensive understanding of the role of T cells in the ageing brain and the underlying molecular pathways under normal conditions could pave the way for new research to better understand brain disorders. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
| | - Raquel Sánchez-Díaz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | | | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
12
|
Levayer R. Staying away from the breaking point: Probing the limits of epithelial cell elimination. Curr Opin Cell Biol 2024; 86:102316. [PMID: 38199024 DOI: 10.1016/j.ceb.2023.102316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
Epithelial tissues are dramatically remodelled during embryogenesis and tissue homeostasis and yet need to maintain their sealing properties to sustain their barrier functions at any time. Part of these remodellings involve the elimination of a large proportion of cells through apoptosis. Cell extrusion, the remodelling steps leading to seamless dying cell expulsion, helps to maintain tissue cohesion. However, there is an intrinsic limit in the system that can only accommodate a certain proportion/rate of cell elimination as well as certain spatiotemporal distributions. What are then the critical conditions leading to epithelial rupture/tear/sealing defects upon cell elimination and which mechanisms ensure that such limits are never reached? In this short review, I document the conditions in which epithelial rupture has been observed, including in the contexts of epithelial cell death, and the mechanical parameters influencing tissue rupture, and review feedback mechanisms which help to keep the epithelia away from the breaking point.
Collapse
Affiliation(s)
- Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
13
|
McCloskey MC, Kasap P, Trempel M, Widom LP, Kuebel J, Chen K, Gaborski TR, Engelhardt B, McGrath JL. Use of the MicroSiM (µSiM) Barrier Tissue Platform for Modeling the Blood-Brain Barrier. J Vis Exp 2024:10.3791/65258. [PMID: 38284519 PMCID: PMC11293877 DOI: 10.3791/65258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024] Open
Abstract
The microSiM (µSiM) is a membrane-based culture platform for modeling the blood-brain barrier (BBB). Unlike conventional membrane-based platforms, the µSiM provides experimentalists with new capabilities, including live cell imaging, unhindered paracrine signaling between 'blood' and 'brain' chambers, and the ability to directly image immunofluorescence without the need for the extraction/remounting of membranes. Here we demonstrate the basic use of the platform to establish monoculture (endothelial cells) and co-culture (endothelial cells and pericytes) models of the BBB using ultrathin nanoporous silicon-nitride membranes. We demonstrate compatibility with both primary cell cultures and human induced pluripotent stem cell (hiPSC) cultures. We provide methods for qualitative analysis of BBB models via immunofluorescence staining and demonstrate the use of the µSiM for the quantitative assessment of barrier function in a small molecule permeability assay. The methods provided should enable users to establish their barrier models on the platform, advancing the use of tissue chip technology for studying human tissues.
Collapse
Affiliation(s)
| | - Pelin Kasap
- Theodor Kocher Institute, University of Bern
| | | | - Louis P Widom
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Julia Kuebel
- Department of Biomedical Engineering, University of Rochester
| | - Kaihua Chen
- Department of Biomedical Engineering, University of Rochester
| | - Thomas R Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology
| | | | - James L McGrath
- Department of Biomedical Engineering, University of Rochester;
| |
Collapse
|
14
|
Lyck R, Nishihara H, Aydin S, Soldati S, Engelhardt B. Modeling Brain Vasculature Immune Interactions In Vitro. Cold Spring Harb Perspect Med 2023; 13:a041185. [PMID: 36617644 PMCID: PMC10513158 DOI: 10.1101/cshperspect.a041185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The endothelial blood-brain barrier (BBB) protects central nervous system (CNS) neurons from the changeable milieu of the bloodstream by strictly controlling the movement of molecules and immune cells between the blood and the CNS. Immune cell migration across the vascular wall is a multistep process regulated by the sequential interaction of different signaling and adhesion molecules on the endothelium and the immune cells. Accounting for its unique barrier properties and trafficking molecule expression profile, particular adaptions in immune cell migration across the BBB have been observed. Thus, in vitro models of the BBB are desirable to explore the precise cellular and molecular mechanisms involved in immune cell trafficking across the BBB. The challenge to overcome is that barrier properties of brain microvascular endothelial cells are not intrinsic and readily lost in culture. With a focus on human in vitro BBB models, we here discuss the suitability of available in vitro models for the BBB for exploring the specific mechanisms involved in immune cell trafficking across the BBB.
Collapse
Affiliation(s)
- Ruth Lyck
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Sidar Aydin
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| |
Collapse
|
15
|
Yan L, Dwiggins CW, Moriarty RA, Jung JW, Gupta U, Brandon KD, Stroka KM. Matrix stiffness regulates the tight junction phenotypes and local barrier properties in tricellular regions in an iPSC-derived BBB model. Acta Biomater 2023:S1742-7061(23)00327-6. [PMID: 37302732 DOI: 10.1016/j.actbio.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
The blood-brain barrier (BBB) can respond to various mechanical cues such as shear stress and substrate stiffness. In the human brain, the compromised barrier function of the BBB is closely associated with a series of neurological disorders that are often also accompanied by the alteration of brain stiffness. In many types of peripheral vasculature, higher matrix stiffness decreases barrier function of endothelial cells through mechanotransduction pathways that alter cell-cell junction integrity. However, human brain endothelial cells are specialized endothelial cells that largely resist changes in cell morphology and key BBB markers. Therefore, it has remained an open question how matrix stiffness affects barrier integrity in the human BBB. To gain insight into the effects of matrix stiffness on BBB permeability, we differentiated brain microvascular endothelial-like cells from human induced pluripotent stem cells (iBMEC-like cells) and cultured the cells on extracellular matrix-coated hydrogels of varying stiffness. We first detected and quantified the junction presentation of key tight junction (TJ) proteins. Our results show matrix-dependent junction phenotypes in iBMEC-like cells, where cells on softer gels (1 kPa) have significantly lower continuous and total TJ coverages. We also determined that these softer gels also lead to decreased barrier function in a local permeability assay. Furthermore, we found that matrix stiffness regulates the local permeability of iBMEC-like cells through the balance of continuous ZO-1 TJs and no junction regions ZO-1 in tricellular regions. Together, these findings provide valuable insights into the effects of matrix stiffness on TJ phenotypes and local permeability of iBMEC-like cells. STATEMENT OF SIGNIFICANCE: Brain mechanical properties, including stiffness, are particularly sensitive indicators for pathophysiological changes in neural tissue. The compromised function of the blood-brain barrier is closely associated with a series of neurological disorders often accompanied by altered brain stiffness. In this study, we use polymeric biomaterials and provide new evidence that biomaterial stiffness regulates the local permeability in iPSC-derived brain endothelial cells in tricellular regions through the tight junction protein ZO-1. Our findings provide valuable insights into the changes in junction architecture and barrier permeability in response to different substrate stiffnesses. Since BBB dysfunction has been linked to many diseases, understanding the influence of substrate stiffness on junction presentations and barrier permeability could lead to the development of new treatments for diseases associated with BBB dysfunction or drug delivery across BBB systems.
Collapse
Affiliation(s)
- Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
| | - Cole W Dwiggins
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Rebecca A Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Udit Gupta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Ken D Brandon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Biophysics Program, University of Maryland, College Park, MD 20742, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
16
|
Haydinger CD, Ashander LM, Tan ACR, Smith JR. Intercellular Adhesion Molecule 1: More than a Leukocyte Adhesion Molecule. BIOLOGY 2023; 12:biology12050743. [PMID: 37237555 DOI: 10.3390/biology12050743] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a transmembrane protein in the immunoglobulin superfamily expressed on the surface of multiple cell populations and upregulated by inflammatory stimuli. It mediates cellular adhesive interactions by binding to the β2 integrins macrophage antigen 1 and leukocyte function-associated antigen 1, as well as other ligands. It has important roles in the immune system, including in leukocyte adhesion to the endothelium and transendothelial migration, and at the immunological synapse formed between lymphocytes and antigen-presenting cells. ICAM-1 has also been implicated in the pathophysiology of diverse diseases from cardiovascular diseases to autoimmune disorders, certain infections, and cancer. In this review, we summarize the current understanding of the structure and regulation of the ICAM1 gene and the ICAM-1 protein. We discuss the roles of ICAM-1 in the normal immune system and a selection of diseases to highlight the breadth and often double-edged nature of its functions. Finally, we discuss current therapeutics and opportunities for advancements.
Collapse
Affiliation(s)
- Cameron D Haydinger
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Liam M Ashander
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Alwin Chun Rong Tan
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Justine R Smith
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| |
Collapse
|
17
|
Modvig S, Jeyakumar J, Marquart HV, Christensen C. Integrins and the Metastasis-like Dissemination of Acute Lymphoblastic Leukemia to the Central Nervous System. Cancers (Basel) 2023; 15:cancers15092504. [PMID: 37173970 PMCID: PMC10177281 DOI: 10.3390/cancers15092504] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) disseminates with high prevalence to the central nervous system (CNS) in a process resembling aspects of the CNS surveillance of normal immune cells as well as aspects of brain metastasis from solid cancers. Importantly, inside the CNS, the ALL blasts are typically confined within the cerebrospinal fluid (CSF)-filled cavities of the subarachnoid space, which they use as a sanctuary protected from both chemotherapy and immune cells. At present, high cumulative doses of intrathecal chemotherapy are administered to patients, but this is associated with neurotoxicity and CNS relapse still occurs. Thus, it is imperative to identify markers and novel therapy targets specific to CNS ALL. Integrins represent a family of adhesion molecules involved in cell-cell and cell-matrix interactions, implicated in the adhesion and migration of metastatic cancer cells, normal immune cells, and leukemic blasts. The ability of integrins to also facilitate cell-adhesion mediated drug resistance, combined with recent discoveries of integrin-dependent routes of leukemic cells into the CNS, have sparked a renewed interest in integrins as markers and therapeutic targets in CNS leukemia. Here, we review the roles of integrins in CNS surveillance by normal lymphocytes, dissemination to the CNS by ALL cells, and brain metastasis from solid cancers. Furthermore, we discuss whether ALL dissemination to the CNS abides by known hallmarks of metastasis, and the potential roles of integrins in this context.
Collapse
Affiliation(s)
- Signe Modvig
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jenani Jeyakumar
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Claus Christensen
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
18
|
Zakani M, Nigritinou M, Ponleitner M, Takai Y, Hofmann D, Hillebrand S, Höftberger R, Bauer J, Lasztoczi B, Misu T, Kasprian G, Rommer P, Bradl M. Paths to hippocampal damage in neuromyelitis optica spectrum disorders. Neuropathol Appl Neurobiol 2023; 49:e12893. [PMID: 36811295 PMCID: PMC10947283 DOI: 10.1111/nan.12893] [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: 07/21/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
AIMS Many patients with neuromyelitis optica spectrum disorders (NMOSD) suffer from cognitive impairment affecting memory, processing speed and attention and suffer from depressive symptoms. Because some of these manifestations could trace back to the hippocampus, several magnetic resonance imaging (MRI) studies have been performed in the past, with a number of groups describing volume loss of the hippocampus in NMOSD patients, whereas others did not observe such changes. Here, we addressed these discrepancies. METHODS We performed pathological and MRI studies on the hippocampi of NMOSD patients, combined with detailed immunohistochemical analysis of hippocampi from experimental models of NMOSD. RESULTS We identified different pathological scenarios for hippocampal damage in NMOSD and its experimental models. In the first case, the hippocampus was compromised by the initiation of astrocyte injury in this brain region and subsequent local effects of microglial activation and neuronal damage. In the second case, loss of hippocampal volume was seen by MRI in patients with large tissue-destructive lesions in the optic nerves or the spinal cord, and the pathological work-up of tissue derived from a patient with such lesions revealed subsequent retrograde neuronal degeneration affecting different axonal tracts and neuronal networks. It remains to be seen whether remote lesions and associated retrograde neuronal degeneration on their own are sufficient to cause extensive volume loss of the hippocampus, or whether they act in concert with small astrocyte-destructive, microglia-activating lesions in the hippocampus that escape detection by MRI, either due to their small size or due to the chosen time window for examination. CONCLUSIONS Different pathological scenarios can culminate in hippocampal volume loss in NMOSD patients.
Collapse
Affiliation(s)
- Mona Zakani
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Magdalini Nigritinou
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | | | - Yoshiki Takai
- Department of NeurologyTohoku University Graduate School of MedicineSendaiJapan
| | - Daniel Hofmann
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Sophie Hillebrand
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Romana Höftberger
- Department of Neurology, Division of Neuropathology and NeurochemistryMedical University of ViennaViennaAustria
| | - Jan Bauer
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Balint Lasztoczi
- Division of Cognitive Neurobiology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Tatsuro Misu
- Department of NeurologyTohoku University Graduate School of MedicineSendaiJapan
| | - Gregor Kasprian
- Division of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Paulus Rommer
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Monika Bradl
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| |
Collapse
|
19
|
Hudecz D, McCloskey MC, Vergo S, Christensen S, McGrath JL, Nielsen MS. Modelling a Human Blood-Brain Barrier Co-Culture Using an Ultrathin Silicon Nitride Membrane-Based Microfluidic Device. Int J Mol Sci 2023; 24:5624. [PMID: 36982697 PMCID: PMC10058651 DOI: 10.3390/ijms24065624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Understanding the vesicular trafficking of receptors and receptor ligands in the brain capillary endothelium is essential for the development of the next generations of biologics targeting neurodegenerative diseases. Such complex biological questions are often approached by in vitro models in combination with various techniques. Here, we present the development of a stem cell-based human in vitro blood-brain barrier model composed of induced brain microvascular endothelial cells (iBMECs) on the modular µSiM (a microdevice featuring a silicon nitride membrane) platform. The µSiM was equipped with a 100 nm thick nanoporous silicon nitride membrane with glass-like imaging quality that allowed the use of high-resolution in situ imaging to study the intracellular trafficking. As a proof-of-concept experiment, we investigated the trafficking of two monoclonal antibodies (mAb): an anti-human transferrin receptor mAb (15G11) and an anti-basigin mAb (#52) using the µSiM-iBMEC-human astrocyte model. Our results demonstrated effective endothelial uptake of the selected antibodies; however, no significant transcytosis was observed when the barrier was tight. In contrast, when the iBMECs did not form a confluent barrier on the µSiM, the antibodies accumulated inside both the iBMECs and astrocytes, demonstrating that the cells have an active endocytic and subcellular sorting machinery and that the µSiM itself does not hinder antibody transport. In conclusion, our µSiM-iBMEC-human astrocyte model provides a tight barrier with endothelial-like cells, which can be used for high-resolution in situ imaging and for studying receptor-mediated transport and transcytosis in a physiological barrier.
Collapse
Affiliation(s)
- Diana Hudecz
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Sandra Vergo
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| | - Søren Christensen
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | | |
Collapse
|
20
|
Su SH, Song Y, Stephens A, Situ M, McCloskey MC, McGrath JL, Andjelkovic AV, Singer BH, Kurabayashi K. A tissue chip with integrated digital immunosensors: In situ brain endothelial barrier cytokine secretion monitoring. Biosens Bioelectron 2023; 224:115030. [PMID: 36603283 PMCID: PMC10401069 DOI: 10.1016/j.bios.2022.115030] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Organ-on-a-chip platforms have potential to offer more cost-effective, ethical, and human-resembling models than animal models for disease study and drug discovery. Particularly, the Blood-Brain-Barrier-on-a-chip (BBB-oC) has emerged as a promising tool to investigate several neurological disorders since it promises to provide a model of the multifunctional tissue working as an important node to control pathogen entry, drug delivery and neuroinflammation. A comprehensive understanding of the multiple physiological functions of the tissue model requires biosensors detecting several tissue-secreted substances in a BBB-oC system. However, current sensor-integrated BBB-oC platforms are only available for tissue membrane integrity characterization based on permeability measurement. Protein secretory pathways are closely associated with the tissue's various diseased conditions. At present, no biosensor-integrated BBB-oC platform exists that permits in situ tissue protein secretion analysis over time, which prohibits researchers from fully understanding the time-evolving pathology of a tissue barrier. Herein, the authors present a platform named "Digital Tissue-BArrier-CytoKine-counting-on-a-chip (DigiTACK)," which integrates digital immunosensors into a tissue chip system and demonstrates on-chip multiplexed, ultrasensitive, longitudinal cytokine secretion profiling of cultured brain endothelial barrier tissues. The integrated digital sensors utilize a novel beadless microwell format to perform an ultrafast "digital fingerprinting" of the analytes while achieving a low limit of detection (LoD) around 100-500 fg/mL for mouse MCP1 (CCL2), IL-6 and KC (CXCL1). The DigiTACK platform is extensively applicable to profile temporal cytokine secretion of other barrier-related organ-on-a-chip systems and can provide new insight into the secretory dynamics of the BBB by sequentially controlled experiments.
Collapse
Affiliation(s)
- Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew Stephens
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Muyu Situ
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Molly C McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Anuska V Andjelkovic
- Department of Pathology and Neurosurgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin H Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
21
|
Harris WJ, Asselin MC, Hinz R, Parkes LM, Allan S, Schiessl I, Boutin H, Dickie BR. In vivo methods for imaging blood-brain barrier function and dysfunction. Eur J Nucl Med Mol Imaging 2023; 50:1051-1083. [PMID: 36437425 PMCID: PMC9931809 DOI: 10.1007/s00259-022-05997-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/09/2022] [Indexed: 11/29/2022]
Abstract
The blood-brain barrier (BBB) is the interface between the central nervous system and systemic circulation. It tightly regulates what enters and is removed from the brain parenchyma and is fundamental in maintaining brain homeostasis. Increasingly, the BBB is recognised as having a significant role in numerous neurological disorders, ranging from acute disorders (traumatic brain injury, stroke, seizures) to chronic neurodegeneration (Alzheimer's disease, vascular dementia, small vessel disease). Numerous approaches have been developed to study the BBB in vitro, in vivo, and ex vivo. The complex multicellular structure and effects of disease are difficult to recreate accurately in vitro, and functional aspects of the BBB cannot be easily studied ex vivo. As such, the value of in vivo methods to study the intact BBB cannot be overstated. This review discusses the structure and function of the BBB and how these are affected in diseases. It then discusses in depth several established and novel methods for imaging the BBB in vivo, with a focus on MRI, nuclear imaging, and high-resolution intravital fluorescence microscopy.
Collapse
Affiliation(s)
- William James Harris
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Marie-Claude Asselin
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Laura Michelle Parkes
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Stuart Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Herve Boutin
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
| | - Ben Robert Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
22
|
Grönloh MLB, Arts JJG, Palacios Martínez S, van der Veen AA, Kempers L, van Steen ACI, Roelofs JJTH, Nolte MA, Goedhart J, van Buul JD. Endothelial transmigration hotspots limit vascular leakage through heterogeneous expression of ICAM-1. EMBO Rep 2023; 24:e55483. [PMID: 36382783 PMCID: PMC9827561 DOI: 10.15252/embr.202255483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Upon inflammation, leukocytes leave the circulation by crossing the endothelial monolayer at specific transmigration "hotspot" regions. Although these regions support leukocyte transmigration, their functionality is not clear. We found that endothelial hotspots function to limit vascular leakage during transmigration events. Using the photoconvertible probe mEos4b, we traced back and identified original endothelial transmigration hotspots. Using this method, we show that the heterogeneous distribution of ICAM-1 determines the location of the transmigration hotspot. Interestingly, the loss of ICAM-1 heterogeneity either by CRISPR/Cas9-induced knockout of ICAM-1 or equalizing the distribution of ICAM-1 in all endothelial cells results in the loss of TEM hotspots but not necessarily in reduced TEM events. Functionally, the loss of endothelial hotspots results in increased vascular leakage during TEM. Mechanistically, we demonstrate that the 3 extracellular Ig-like domains of ICAM-1 are crucial for hotspot recognition. However, the intracellular tail of ICAM-1 and the 4th Ig-like dimerization domain are not involved, indicating that intracellular signaling or ICAM-1 dimerization is not required for hotspot recognition. Together, we discovered that hotspots function to limit vascular leakage during inflammation-induced extravasation.
Collapse
Affiliation(s)
- Max L B Grönloh
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
- Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced MicroscopyUniversity of AmsterdamAmsterdamThe Netherlands
| | - Janine J G Arts
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
- Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced MicroscopyUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sebastián Palacios Martínez
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
| | - Amerens A van der Veen
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
| | - Lanette Kempers
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
| | - Abraham C I van Steen
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Location AMCAmsterdamThe Netherlands
| | - Martijn A Nolte
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
| | - Joachim Goedhart
- Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced MicroscopyUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab, Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
- Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced MicroscopyUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
23
|
Mansouri M, Ahmed A, Ahmad SD, McCloskey MC, Joshi IM, Gaborski TR, Waugh RE, McGrath JL, Day SW, Abhyankar VV. The Modular µSiM Reconfigured: Integration of Microfluidic Capabilities to Study In Vitro Barrier Tissue Models under Flow. Adv Healthc Mater 2022; 11:e2200802. [PMID: 35953453 PMCID: PMC9798530 DOI: 10.1002/adhm.202200802] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/01/2022] [Indexed: 01/28/2023]
Abstract
Microfluidic tissue barrier models have emerged to address the lack of physiological fluid flow in conventional "open-well" Transwell-like devices. However, microfluidic techniques have not achieved widespread usage in bioscience laboratories because they are not fully compatible with traditional experimental protocols. To advance barrier tissue research, there is a need for a platform that combines the key advantages of both conventional open-well and microfluidic systems. Here, a plug-and-play flow module is developed to introduce on-demand microfluidic flow capabilities to an open-well device that features a nanoporous membrane and live-cell imaging capabilities. The magnetic latching assembly of this design enables bi-directional reconfiguration and allows users to conduct an experiment in an open-well format with established protocols and then add or remove microfluidic capabilities as desired. This work also provides an experimentally-validated flow model to select flow conditions based on the experimental needs. As a proof-of-concept, flow-induced alignment of endothelial cells and the expression of shear-sensitive gene targets are demonstrated, and the different phases of neutrophil transmigration across a chemically stimulated endothelial monolayer under flow conditions are visualized. With these experimental capabilities, it is anticipated that both engineering and bioscience laboratories will adopt this reconfigurable design due to the compatibility with standard open-well protocols.
Collapse
Affiliation(s)
- Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Indranil M. Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Thomas R. Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Richard E. Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Steven W. Day
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Vinay V. Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| |
Collapse
|
24
|
Williams DW, Flores BR, Xu Y, Wang Y, Yu D, Peters BA, Adedimeji A, Wilson TE, Merenstein D, Tien PC, Cohen MH, Weber KM, Adimora AA, Ofotokun I, Fischl M, Turan J, Turan B, Laumet G, Landay AL, Dastgheyb RM, Gange SJ, Weiser SD, Rubin LH. T-cell activation state differentially contributes to neuropsychiatric complications in women with HIV. Brain Behav Immun Health 2022; 25:100498. [PMID: 36097532 PMCID: PMC9463560 DOI: 10.1016/j.bbih.2022.100498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/07/2022] [Accepted: 08/13/2022] [Indexed: 02/02/2023] Open
Abstract
Neuropsychiatric complications are common among women with HIV (WWH). The pathophysiological mechanisms underlying these complications are not fully known but likely driven in part by immune modulation. We examined associations between T-cell activation states which are required to mount an effective immune response (activation, co-stimulation/normal function, exhaustion, senescence) and neuropsychiatric complications in WWH. 369 WWH (78% HIV RNA undetectable/<20cp/mL) enrolled in the Women's Interagency HIV Study completed neuropsychological testing and measures of depression (Center for Epidemiological Studies Depression Scale-CES-D), self-reported stress levels (Perceived Stress Scale-10), and post-traumatic stress (PTSD Checklist-Civilian Scale). Multiparametric flow cytometry evaluated T-cell activation state. Partial least squares regressions were used to examine T-cell phenotypes and neuropsychiatric outcome associations after confounder adjustment. In the total sample and among virally suppressed (VS)-WWH, CD4+ T-cell exhaustion was associated with poorer learning and attention/working memory (P's < 0.05). In the total sample, CD4+ T-cell activation was associated with better attention/working memory and CD8+ T-cell co-stimulation and senescence was associated with poorer executive function (P's < 0.05). For mental health outcomes, in the total sample, CD4+ T-cell activation was associated with more perceived stress and CD4+ T-cell exhaustion was associated with less depressive symptoms (P's < 0.05). Among VS-WWH, CD4+ senescence was associated with less perceive stress and CD8+ T-cell co-stimulation and senescence was associated with higher depression (P's < 0.05). Together, results suggest the contribution of peripheral CD4+ and CD8+ T-cell activation status to neuropsychiatric complications in WWH.
Collapse
Affiliation(s)
- Dionna W. Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bianca R. Flores
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yanxun Xu
- Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
- Division of Biostatistics and Bioinformatics at the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuezhe Wang
- Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Danyang Yu
- Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Brandilyn A. Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adebola Adedimeji
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tracey E. Wilson
- Department of Community Health Sciences, State University of New York Downstate Health Science University, School of Public Health, Brooklyn, NY, USA
| | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Phyllis C. Tien
- Department of Medicine, UCSF and Medical Service, Department of Veteran Affairs Medical Center, San Francisco, CA, USA
| | | | | | - Adaora A. Adimora
- Division of Infectious Disease, University of North Carolina at Chapel Hill, NC, USA
| | - Igho Ofotokun
- Department of Medicine, Emory University and Grady Healthcare System, Atlanta, Georgia Mailman School of Public Health, Columbia University, NY, NY, USA
| | - Margaret Fischl
- Department of Medicine, University of Miami Health System, Miami, FL, USA
| | - Janet Turan
- Departments of Health Policy and Organization, School of Public Health, University of Alabama at Birmingham, USA
| | - Bülent Turan
- Department of Psychology, Koc University, Istanbul, Turkey
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Raha M. Dastgheyb
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen J. Gange
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sheri D. Weiser
- Department of Medicine, UCSF and Medical Service, Department of Veteran Affairs Medical Center, San Francisco, CA, USA
- Division of HIV, ID and Global Medicine, Department of Medicine, UCSF, San Francisco, CA, USA
| | - Leah H. Rubin
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Goncalves A, Antonetti DA. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids Barriers CNS 2022; 19:86. [PMID: 36320068 PMCID: PMC9628113 DOI: 10.1186/s12987-022-00386-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
The unique environment of the brain and retina is tightly regulated by blood-brain barrier and the blood-retinal barrier, respectively, to ensure proper neuronal function. Endothelial cells within these tissues possess distinct properties that allow for controlled passage of solutes and fluids. Pericytes, glia cells and neurons signal to endothelial cells (ECs) to form and maintain the barriers and control blood flow, helping to create the neurovascular unit. This barrier is lost in a wide range of diseases affecting the central nervous system (CNS) and retina such as brain tumors, stroke, dementia, and in the eye, diabetic retinopathy, retinal vein occlusions and age-related macular degeneration to name prominent examples. Recent studies directly link barrier changes to promotion of disease pathology and degradation of neuronal function. Understanding how these barriers form and how to restore these barriers in disease provides an important point for therapeutic intervention. This review aims to describe the fundamentals of the blood-tissue barriers of the CNS and how the use of transgenic animal models led to our current understanding of the molecular framework of these barriers. The review also highlights examples of targeting barrier properties to protect neuronal function in disease states.
Collapse
Affiliation(s)
- Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA.
| |
Collapse
|
26
|
Smith BC, Tinkey RA, Shaw BC, Williams JL. Targetability of the neurovascular unit in inflammatory diseases of the central nervous system. Immunol Rev 2022; 311:39-49. [PMID: 35909222 PMCID: PMC9489669 DOI: 10.1111/imr.13121] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) is a selectively permeable barrier separating the periphery from the central nervous system (CNS). The BBB restricts the flow of most material into and out of the CNS, including many drugs that could be used as potent therapies. BBB permeability is modulated by several cells that are collectively called the neurovascular unit (NVU). The NVU consists of specialized CNS endothelial cells (ECs), pericytes, astrocytes, microglia, and neurons. CNS ECs maintain a complex "seal" via tight junctions, forming the BBB; breakdown of these tight junctions leads to BBB disruption. Pericytes control the vascular flow within capillaries and help maintain the basal lamina. Astrocytes control much of the flow of material that has moved beyond the CNS EC layer and can form a secondary barrier under inflammatory conditions. Microglia survey the border of the NVU for noxious material. Neuronal activity also plays a role in the maintenance of the BBB. Since astrocytes, pericytes, microglia, and neurons are all able to modulate the permeability of the BBB, understating the complex contributions of each member of the NVU will potentially uncover novel and effective methods for delivery of neurotherapies to the CNS.
Collapse
Affiliation(s)
- Brandon C. Smith
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Department of Biological, Geological, and Environmental SciencesCleveland State UniversityClevelandOhioUSA
| | - Rachel A. Tinkey
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,School of Biomedical SciencesKent State UniversityKentOhioUSA
| | - Benjamin C. Shaw
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Jessica L. Williams
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Brain Health Research Institute, Kent State UniversityKentOhioUSA
| |
Collapse
|
27
|
Quintana JF, Chandrasegaran P, Sinton MC, Briggs EM, Otto TD, Heslop R, Bentley-Abbot C, Loney C, de Lecea L, Mabbott NA, MacLeod A. Single cell and spatial transcriptomic analyses reveal microglia-plasma cell crosstalk in the brain during Trypanosoma brucei infection. Nat Commun 2022; 13:5752. [PMID: 36180478 PMCID: PMC9525673 DOI: 10.1038/s41467-022-33542-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Human African trypanosomiasis, or sleeping sickness, is caused by the protozoan parasite Trypanosoma brucei and induces profound reactivity of glial cells and neuroinflammation when the parasites colonise the central nervous system. However, the transcriptional and functional responses of the brain to chronic T. brucei infection remain poorly understood. By integrating single cell and spatial transcriptomics of the mouse brain, we identify that glial responses triggered by infection are readily detected in the proximity to the circumventricular organs, including the lateral and 3rd ventricle. This coincides with the spatial localisation of both slender and stumpy forms of T. brucei. Furthermore, in silico predictions and functional validations led us to identify a previously unknown crosstalk between homeostatic microglia and Cd138+ plasma cells mediated by IL-10 and B cell activating factor (BAFF) signalling. This study provides important insights and resources to improve understanding of the molecular and cellular responses in the brain during infection with African trypanosomes.
Collapse
Affiliation(s)
- Juan F Quintana
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK.
| | - Praveena Chandrasegaran
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Matthew C Sinton
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Emma M Briggs
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Thomas D Otto
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Infection and Immunity, MVLS, University of Glasgow, Glasgow, UK
| | - Rhiannon Heslop
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Calum Bentley-Abbot
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| | - Colin Loney
- School of Infection and Immunity, MVLS, University of Glasgow, Glasgow, UK
- MRC Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Luis de Lecea
- Stanford University School of Medicine, Stanford, CA, USA
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), MVLS, University of Glasgow, Glasgow, UK
| |
Collapse
|
28
|
Beier LS, Waldow A, Khomeijani Farahani S, Mannweiler R, Vidal-Y-Sy S, Brandner JM, Piontek J, Günzel D. Claudin targeting as an effective tool for directed barrier modulation of the viable epidermis. Ann N Y Acad Sci 2022; 1517:251-265. [PMID: 35994210 DOI: 10.1111/nyas.14879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tight junction (TJ) formation is vital for epidermal barrier function. We aimed to specifically manipulate TJ barriers in the reconstructed human epidermis (RHE) by claudin-1 and -4 knockdown (KD) and by claudin-binding fusion proteins of glutathione S-transferase and modified C-terminal fragments of Clostridium perfringens enterotoxin (GST-cCPE). Impedance spectroscopy and tracer permeability imaging were employed for functional barrier assessment and investigation of claudin contribution. KD of claudin-1, but not claudin-4, impaired the paracellular barrier in vitro. Similarly, claudin-binding GST-cCPE variants weakened the paracellular but not the stratum corneum barrier. Combining both TJ targeting methods, we found that claudin-1 targeting by GST-cCPE after claudin-4 KD led to a marked decrease in paracellular barrier properties. Conversely, after claudin-1 KD, GST-cCPE did not further impair the barrier. Comparison of GST-cCPE variants with different claudin-1/claudin-4 affinities, NHS-fluorescein tracer detection, and immunostaining of RHE paraffin sections showed that GST-cCPE variants bind to extrajunctional claudin-1 and -4, which are differentially distributed along the stratum basale-stratum granulosum axis. GST-cCPE binding blocks these claudins, thereby specifically opening the paracellular barrier of RHE. The data indicate a critical role for claudin-1 in regulating paracellular permeability for ions and small molecules in the viable epidermis. Claudin targeting is presented as a proof-of-concept for precise barrier modulation.
Collapse
Affiliation(s)
- Laura-Sophie Beier
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ayk Waldow
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Saeed Khomeijani Farahani
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roman Mannweiler
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sabine Vidal-Y-Sy
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna M Brandner
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Piontek
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
29
|
Barkaway A, Attwell D, Korte N. Immune-vascular mural cell interactions: consequences for immune cell trafficking, cerebral blood flow, and the blood-brain barrier. NEUROPHOTONICS 2022; 9:031914. [PMID: 35581998 PMCID: PMC9107322 DOI: 10.1117/1.nph.9.3.031914] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 06/15/2023]
Abstract
Brain barriers are crucial sites for cerebral energy supply, waste removal, immune cell migration, and solute exchange, all of which maintain an appropriate environment for neuronal activity. At the capillary level, where the largest area of brain-vascular interface occurs, pericytes adjust cerebral blood flow (CBF) by regulating capillary diameter and maintain the blood-brain barrier (BBB) by suppressing endothelial cell (EC) transcytosis and inducing tight junction expression between ECs. Pericytes also limit the infiltration of circulating leukocytes into the brain where resident microglia confine brain injury and provide the first line of defence against invading pathogens. Brain "waste" is cleared across the BBB into the blood, phagocytosed by microglia and astrocytes, or removed by the flow of cerebrospinal fluid (CSF) through perivascular routes-a process driven by respiratory motion and the pulsation of the heart, arteriolar smooth muscle, and possibly pericytes. "Dirty" CSF exits the brain and is probably drained around olfactory nerve rootlets and via the dural meningeal lymphatic vessels and possibly the skull bone marrow. The brain is widely regarded as an immune-privileged organ because it is accessible to few antigen-primed leukocytes. Leukocytes enter the brain via the meninges, the BBB, and the blood-CSF barrier. Advances in genetic and imaging tools have revealed that neurological diseases significantly alter immune-brain barrier interactions in at least three ways: (1) the brain's immune-privileged status is compromised when pericytes are lost or lymphatic vessels are dysregulated; (2) immune cells release vasoactive molecules to regulate CBF, modulate arteriole stiffness, and can plug and eliminate capillaries which impairs CBF and possibly waste clearance; and (3) immune-vascular interactions can make the BBB leaky via multiple mechanisms, thus aggravating the influx of undesirable substances and cells. Here, we review developments in these three areas and briefly discuss potential therapeutic avenues for restoring brain barrier functions.
Collapse
Affiliation(s)
- Anna Barkaway
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - David Attwell
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Nils Korte
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| |
Collapse
|
30
|
Proulx ST, Engelhardt B. Central nervous system zoning: How brain barriers establish subdivisions for CNS immune privilege and immune surveillance. J Intern Med 2022; 292:47-67. [PMID: 35184353 PMCID: PMC9314672 DOI: 10.1111/joim.13469] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The central nervous system (CNS) coordinates all our body functions. Neurons in the CNS parenchyma achieve this computational task by high speed communication via electrical and chemical signals and thus rely on a strictly regulated homeostatic environment, which does not tolerate uncontrolled entry of blood components including immune cells. The CNS thus has a unique relationship with the immune system known as CNS immune privilege. Previously ascribed to the presence of blood-brain barriers and the lack of lymphatic vessels in the CNS parenchyma prohibiting, respectively, efferent and afferent connections with the peripheral immune system, it is now appreciated that CNS immune surveillance is ensured by cellular and acellular brain barriers that limit immune cell and mediator accessibility to specific compartments at the borders of the CNS. CNS immune privilege is established by a brain barriers anatomy resembling the architecture of a medieval castle surrounded by two walls bordering a castle moat. Built for protection and defense this two-walled rampart at the outer perimeter of the CNS parenchyma allows for accommodation of different immune cell subsets and efficient monitoring of potential danger signals derived from inside or outside of the CNS parenchyma. It enables effective mounting of immune responses within the subarachnoid or perivascular spaces, while leaving the CNS parenchyma relatively undisturbed. In this study, we propose that CNS immune privilege rests on the proper function of the brain barriers, which allow for CNS immune surveillance but prohibit activation of immune responses from the CNS parenchyma unless it is directly injured.
Collapse
Affiliation(s)
- Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
31
|
Keep RF, Jones HC, Drewes LR. Advances in brain barriers and brain fluids research in 2021: great progress in a time of adversity. Fluids Barriers CNS 2022; 19:48. [PMID: 35681151 PMCID: PMC9178944 DOI: 10.1186/s12987-022-00343-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
This editorial highlights advances in brain barrier and brain fluid research in 2021. It covers research on components of the blood–brain barrier, neurovascular unit and brain fluid systems; how brain barriers and brain fluid systems are impacted by neurological disorders and their role in disease progression; and advances in strategies for treating such disorders.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | | | - Lester R Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN, 55812, USA
| |
Collapse
|
32
|
Engelhardt B, Comabella M, Chan A. Multiple sclerosis: Immunopathological heterogeneity and its implications. Eur J Immunol 2022; 52:869-881. [PMID: 35476319 PMCID: PMC9324211 DOI: 10.1002/eji.202149757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 01/13/2023]
Abstract
MS is the most common autoimmune demyelinating disease of the CNS. For the past decades, several immunomodulatory disease-modifying treatments with multiple presumed mechanisms of action have been developed, but MS remains an incurable disease. Whereas high efficacy, at least in early disease, corroborates underlying immunopathophysiology, there is profound heterogeneity in clinical presentation as well as immunophenotypes that may also vary over time. In addition, functional plasticity in the immune system as well as in the inflamed CNS further contributes to disease heterogeneity. In this review, we will highlight immune-pathophysiological and associated clinical heterogeneity that may have an implication for more precise immunomodulatory therapeutic strategies in MS.
Collapse
Affiliation(s)
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Mapunda JA, Tibar H, Regragui W, Engelhardt B. How Does the Immune System Enter the Brain? Front Immunol 2022; 13:805657. [PMID: 35273596 PMCID: PMC8902072 DOI: 10.3389/fimmu.2022.805657] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple Sclerosis (MS) is considered the most frequent inflammatory demyelinating disease of the central nervous system (CNS). It occurs with a variable prevalence across the world. A rich armamentarium of disease modifying therapies selectively targeting specific actions of the immune system is available for the treatment of MS. Understanding how and where immune cells are primed, how they access the CNS in MS and how immunomodulatory treatments affect neuroinflammation requires a proper knowledge on the mechanisms regulating immune cell trafficking and the special anatomy of the CNS. The brain barriers divide the CNS into different compartments that differ with respect to their accessibility to cells of the innate and adaptive immune system. In steady state, the blood-brain barrier (BBB) limits immune cell trafficking to activated T cells, which can reach the cerebrospinal fluid (CSF) filled compartments to ensure CNS immune surveillance. In MS immune cells breach a second barrier, the glia limitans to reach the CNS parenchyma. Here we will summarize the role of the endothelial, epithelial and glial brain barriers in regulating immune cell entry into the CNS and which immunomodulatory treatments for MS target the brain barriers. Finally, we will explore current knowledge on genetic and environmental factors that may influence immune cell entry into the CNS during neuroinflammation in Africa.
Collapse
Affiliation(s)
| | - Houyam Tibar
- Medical School of Rabat, Mohamed 5 University, Rabat, Morocco.,Hôpital des spécialités de Rabat, Ibn Sina University Hospital of Rabat, Rabat, Morocco
| | - Wafa Regragui
- Medical School of Rabat, Mohamed 5 University, Rabat, Morocco.,Hôpital des spécialités de Rabat, Ibn Sina University Hospital of Rabat, Rabat, Morocco
| | | |
Collapse
|
34
|
Pogodalla N, Winkler B, Klämbt C. Glial Tiling in the Insect Nervous System. Front Cell Neurosci 2022; 16:825695. [PMID: 35250488 PMCID: PMC8891220 DOI: 10.3389/fncel.2022.825695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/06/2022] [Indexed: 11/30/2022] Open
Abstract
The Drosophila nervous system comprises a small number of well characterized glial cell classes. The outer surface of the central nervous system (CNS) is protected by a glial derived blood-brain barrier generated by perineurial and subperineurial glia. All neural stem cells and all neurons are engulfed by cortex glial cells. The inner neuropil region, that harbors all synapses and dendrites, is covered by ensheathing glia and infiltrated by astrocyte-like glial cells. All these glial cells show a tiled organization with an often remarkable plasticity where glial cells of one cell type invade the territory of the neighboring glial cell type upon its ablation. Here, we summarize the different glial tiling patterns and based on the different modes of cell-cell contacts we hypothesize that different molecular mechanisms underlie tiling of the different glial cell types.
Collapse
|
35
|
Hudson N, Campbell M. Tight Junctions of the Neurovascular Unit. Front Mol Neurosci 2021; 14:752781. [PMID: 34867185 PMCID: PMC8640090 DOI: 10.3389/fnmol.2021.752781] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
The homeostatic balance of the brain and retina is maintained by the presence of the blood-brain and inner blood-retinal barrier (BBB/iBRB, respectively) which are highly specialized barriers. Endothelial cells forming the lining of these blood vessels are interconnected by the presence of tight junctions which form the BBB and iBRB. These tight junctions, formed of numerous interacting proteins, enable the entry of molecules into neural tissues while restricting the entry of harmful material such as anaphylatoxins, bacteria and viruses. If the tight junction complex becomes dysregulated due to changes in expression levels of one or more of the components, this can have detrimental effects leading to brain and retinal pathology.
Collapse
Affiliation(s)
- Natalie Hudson
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, Ireland
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, Ireland
| |
Collapse
|
36
|
Singh K, Hotchkiss KM, Patel KK, Wilkinson DS, Mohan AA, Cook SL, Sampson JH. Enhancing T Cell Chemotaxis and Infiltration in Glioblastoma. Cancers (Basel) 2021; 13:5367. [PMID: 34771532 PMCID: PMC8582389 DOI: 10.3390/cancers13215367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is an immunologically 'cold' tumor, which are characterized by absent or minimal numbers of tumor-infiltrating lymphocytes (TILs). For those tumors that have been invaded by lymphocytes, they are profoundly exhausted and ineffective. While many immunotherapy approaches seek to reinvigorate immune cells at the tumor, this requires TILs to be present. Therefore, to unleash the full potential of immunotherapy in glioblastoma, the trafficking of lymphocytes to the tumor is highly desirable. However, the process of T cell recruitment into the central nervous system (CNS) is tightly regulated. Naïve T cells may undergo an initial licensing process to enter the migratory phenotype necessary to enter the CNS. T cells then must express appropriate integrins and selectin ligands to interact with transmembrane proteins at the blood-brain barrier (BBB). Finally, they must interact with antigen-presenting cells and undergo further licensing to enter the parenchyma. These T cells must then navigate the tumor microenvironment, which is rich in immunosuppressive factors. Altered tumoral metabolism also interferes with T cell motility. In this review, we will describe these processes and their mediators, along with potential therapeutic approaches to enhance trafficking. We also discuss safety considerations for such approaches as well as potential counteragents.
Collapse
Affiliation(s)
- Kirit Singh
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA; (K.M.H.); (K.K.P.); (D.S.W.); (A.A.M.); (S.L.C.)
| | | | | | | | | | | | - John H. Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA; (K.M.H.); (K.K.P.); (D.S.W.); (A.A.M.); (S.L.C.)
| |
Collapse
|
37
|
Alzheimer's-Like Pathology at the Crossroads of HIV-Associated Neurological Disorders. Vaccines (Basel) 2021; 9:vaccines9080930. [PMID: 34452054 PMCID: PMC8402792 DOI: 10.3390/vaccines9080930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
Despite the widespread success of combined antiretroviral therapy (cART) in suppressing viremia, the prevalence of human immunodeficiency virus (HIV)-associated neurological disorders (HAND) and associated comorbidities such as Alzheimer’s disease (AD)-like symptomatology is higher among people living with HIV. The pathophysiology of observed deficits in HAND is well understood. However, it has been suggested that it is exacerbated by aging. Epidemiological studies have suggested comparable concentrations of the toxic amyloid protein, amyloid-β42 (Aβ42), in the cerebrospinal fluid (CSF) of HAND patients and in the brains of patients with dementia of the Alzheimer’s type. Apart from abnormal amyloid-β (Aβ) metabolism in AD, a better understanding of the role of similar pathophysiologic processes in HAND could be of substantial value. The pathogenesis of HAND involves either the direct effects of the virus or the effect of viral proteins, such as Tat, Gp120, or Nef, as well as the effects of antiretrovirals on amyloid metabolism and tauopathy, leading, in turn, to synaptodendritic alterations and neuroinflammatory milieu in the brain. Additionally, there is a lack of knowledge regarding the causative or bystander role of Alzheimer’s-like pathology in HAND, which is a barrier to the development of therapeutics for HAND. This review attempts to highlight the cause–effect relationship of Alzheimer’s-like pathology with HAND, attempting to dissect the role of HIV-1, HIV viral proteins, and antiretrovirals in patient samples, animal models, and cell culture model systems. Biomarkers associated with Alzheimer’s-like pathology can serve as a tool to assess the neuronal injury in the brain and the associated cognitive deficits. Understanding the factors contributing to the AD-like pathology associated with HAND could set the stage for the future development of therapeutics aimed at abrogating the disease process.
Collapse
|
38
|
First person – Mariana Castro Dias. J Cell Sci 2021. [DOI: 10.1242/jcs.258753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ABSTRACT
First Person is a series of interviews with the first authors of a selection of papers published in Journal of Cell Science, helping early-career researchers promote themselves alongside their papers. Mariana Castro Dias is first author on ‘Brain endothelial tricellular junctions as novel sites for T cell diapedesis across the blood–brain barrier’, published in JCS. Mariana conducted the research described in this article while a PhD student in Britta Engelhardt's lab at the Theodor Kocher Institute, University of Bern, Switzerland. She is now an Associate Scientific Communications Manager working for Roche Diagnostics International. She is utterly fascinated by the brain, particularly the molecular mechanisms underlying the pathology of neurodegenerative disorders.
Collapse
|