1
|
Nogueira Pinto H, Zarekiani P, de Vries HE. Neuroglia and the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:127-141. [PMID: 40122621 DOI: 10.1016/b978-0-443-19104-6.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The blood-brain barrier (BBB) is a highly dynamic and complex structure, present throughout the brain vasculature, that safeguards the brain against blood-borne insults. Neuroglial cells play a major role in its development, function, and homeostasis of the BBB by establishing intricate interactions via direct cell-cell contacts and paracrine signaling. Astrocytes, pericytes, oligodendrocytes, and microglia, alongside specialized brain endothelial cells, orchestrate key events in the brain in health and disease, which can be partially recapitulated by in vitro and in vivo models for biomedical research. This chapter presents a detailed description of the main cellular and molecular mechanisms that govern the neuroglia-BBB crosstalk and the available models for its investigation, emphasizing the importance of each cell population and the synergistic roles they play in the brain.
Collapse
Affiliation(s)
- Henrique Nogueira Pinto
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands
| | - Parand Zarekiani
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Helga E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; MS Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Wakid M, Almeida D, Aouabed Z, Rahimian R, Davoli MA, Yerko V, Leonova-Erko E, Richard V, Zahedi R, Borchers C, Turecki G, Mechawar N. Universal method for the isolation of microvessels from frozen brain tissue: A proof-of-concept multiomic investigation of the neurovasculature. Brain Behav Immun Health 2023; 34:100684. [PMID: 37822873 PMCID: PMC10562768 DOI: 10.1016/j.bbih.2023.100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
The neurovascular unit, comprised of vascular cell types that collectively regulate cerebral blood flow to meet the needs of coupled neurons, is paramount for the proper function of the central nervous system. The neurovascular unit gatekeeps blood-brain barrier properties, which experiences impairment in several central nervous system diseases associated with neuroinflammation and contributes to pathogenesis. To better understand function and dysfunction at the neurovascular unit and how it may confer inflammatory processes within the brain, isolation and characterization of the neurovascular unit is needed. Here, we describe a singular, standardized protocol to enrich and isolate microvessels from archived snap-frozen human and frozen mouse cerebral cortex using mechanical homogenization and centrifugation-separation that preserves the structural integrity and multicellular composition of microvessel fragments. For the first time, microvessels are isolated from postmortem ventromedial prefrontal cortex tissue and are comprehensively investigated as a structural unit using both RNA sequencing and Liquid Chromatography with tandem mass spectrometry (LC-MS/MS). Both the transcriptome and proteome are obtained and compared, demonstrating that the isolated brain microvessel is a robust model for the NVU and can be used to generate highly informative datasets in both physiological and disease contexts.
Collapse
Affiliation(s)
- Marina Wakid
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Zahia Aouabed
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | | | - Volodymyr Yerko
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Elena Leonova-Erko
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Vincent Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - René Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - Christoph Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
3
|
Gschwandtner M, Gammage AN, Deligne C, Mies LFM, Domaingo A, Murdamoothoo D, Loustau T, Schwenzer A, Derler R, Carapito R, Koch M, Mörgelin M, Orend G, Kungl AJ, Midwood KS. Investigating Chemokine-Matrix Networks in Breast Cancer: Tenascin-C Sets the Tone for CCL2. Int J Mol Sci 2023; 24:8365. [PMID: 37176074 PMCID: PMC10179296 DOI: 10.3390/ijms24098365] [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: 03/31/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Bidirectional dialogue between cellular and non-cellular components of the tumor microenvironment (TME) drives cancer survival. In the extracellular space, combinations of matrix molecules and soluble mediators provide external cues that dictate the behavior of TME resident cells. Often studied in isolation, integrated cues from complex tissue microenvironments likely function more cohesively. Here, we study the interplay between the matrix molecule tenascin-C (TNC) and chemokine CCL2, both elevated in and associated with the progression of breast cancer and playing key roles in myeloid immune responses. We uncover a correlation between TNC/CCL2 tissue levels in HER2+ breast cancer and examine the physical and functional interactions of these molecules in a murine disease model with tunable TNC levels and in in vitro cellular and cell-free models. TNC supported sustained CCL2 synthesis, with chemokine binding to TNC via two distinct domains. TNC dominated the behavior of tumor-resident myeloid cells; CCL2 did not impact macrophage survival/activation whilst TNC facilitated an immune suppressive macrophage phenotype that was not dependent on or altered by CCL2 co-expression. Together, these data map new binding partners within the TME and demonstrate that whilst the matrix exerts transcriptional control over the chemokine, each plays a distinct role in subverting anti-tumoral immunity.
Collapse
Affiliation(s)
| | - Anís N. Gammage
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Claire Deligne
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Linda F. M. Mies
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Alissa Domaingo
- Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Devardarssen Murdamoothoo
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 67091 Strasbourg, France
- University of Strasbourg, 67091 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, 67091 Strasbourg, France
| | - Thomas Loustau
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 67091 Strasbourg, France
- University of Strasbourg, 67091 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, 67091 Strasbourg, France
| | - Anja Schwenzer
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Rupert Derler
- Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Raphael Carapito
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, GENOMAX Platform, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, 67091 Strasbourg, France
| | - Manuel Koch
- Institute for Dental Research and Oral, Musculoskeletal Research, Center for Biochemistry, University of Cologne, 50931 Cologne, Germany
| | | | - Gertraud Orend
- INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 67091 Strasbourg, France
- University of Strasbourg, 67091 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67091 Strasbourg, France
- INSERM U1109, The Tumor Microenvironment Group, 67091 Strasbourg, France
| | - Andreas J. Kungl
- Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Kim S. Midwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| |
Collapse
|
4
|
Szpakowski P, Ksiazek-Winiarek D, Czpakowska J, Kaluza M, Milewska-Jedrzejczak M, Glabinski A. Astrocyte-Derived Exosomes Differentially Shape T Cells' Immune Response in MS Patients. Int J Mol Sci 2023; 24:ijms24087470. [PMID: 37108633 PMCID: PMC10138532 DOI: 10.3390/ijms24087470] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Astrocytes, the most abundant group of glia cells in the brain, provide support for neurons and indicate multiple various functions in the central nervous system (CNS). Growing data additionally describe their role in the regulation of immune system activity. They exert their function not only by direct contact with other cell types, but also through an indirect method, e.g., by secreting various molecules. One such structure is extracellular vesicles, which are important mediators of crosstalk between cells. In our study, we observed that the impact of exosomes derived from astrocytes with various functional phenotype differently affect the immune response of CD4+ T cells, both from healthy individuals and from patients with multiple sclerosis (MS). Astrocytes, by modulating exosome cargo, impacts the release of IFN-γ, IL-17A and CCL2 in our experimental conditions. Considering the proteins concentration in cell culture supernatants and the cellular percentage of Th phenotypes, it could be stated that human astrocytes, by the release of exosomes, are able to modify the activity of human T cells.
Collapse
Affiliation(s)
- Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland
| | - Dominika Ksiazek-Winiarek
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland
| | - Joanna Czpakowska
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland
| | - Mateusz Kaluza
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland
| | - Marta Milewska-Jedrzejczak
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland
| | - Andrzej Glabinski
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland
| |
Collapse
|
5
|
Easwaran M, Martinez JD, Kim JB, Erickson-DiRenzo E. Modulation of mouse laryngeal inflammatory and immune cell responses by low and high doses of mainstream cigarette smoke. Sci Rep 2022; 12:18667. [PMID: 36333510 PMCID: PMC9636197 DOI: 10.1038/s41598-022-23359-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Cigarette smoking is a major risk factor for laryngeal diseases. Despite well-documented cigarette smoke (CS) induced laryngeal histopathological changes, the underlying immunopathological mechanisms remain largely unexplored. The goal of this study was to evaluate inflammatory and immune cell responses in a CS-exposed larynx. Specifically, we used a 4-week subacute whole-body CS inhalation mouse model to assess these responses in the laryngeal mucosa upon exposure to low (LD; 1 h/day) and high dose (HD; 4 h/day) CS. Laryngeal tissues were harvested and evaluated using a 254-plex NanoString inflammation panel and neutrophil/macrophage/T-cell immunohistochemistry (IHC). NanoString global and differential gene expression analysis revealed a unique expression profile only in the HD group, with 26 significant differentially expressed genes (DEGs). StringDB KEGG pathway enrichment analysis revealed the involvement of these DEGs with pro-inflammatory pathways including TNF/TNFα and IL-17. Furthermore, inflammatory responses remained inhibited in conjunction with predicted activated states of anti-inflammatory regulators like PPARγ and NFE2L2 upon Ingenuity Pathway Analysis (IPA). Subglottic T-cell levels remained significantly inhibited as corroborated by IPA predictions. Overall, our key findings are consistent with HD exposures being anti-inflammatory and immunosuppressive. Furthermore, the identification of important regulatory genes and enriched pathways may help improve clinical interventions for CS-induced laryngeal diseases.
Collapse
Affiliation(s)
- Meena Easwaran
- Division of Laryngology, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua D Martinez
- Division of Laryngology, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Juyong Brian Kim
- Department of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth Erickson-DiRenzo
- Division of Laryngology, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Craig DJ, Ambrose S, Stanbery L, Walter A, Nemunaitis J. Systemic benefit of radiation therapy via abscopal effect. Front Oncol 2022; 12:987142. [PMID: 36387120 PMCID: PMC9641206 DOI: 10.3389/fonc.2022.987142] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/05/2022] [Indexed: 08/30/2023] Open
Abstract
Evidence of a systemic response related to localized radiation therapy (RT) in cancer management is rare. However, enhancing the immune response via immunotherapy followed by localized RT has shown evidence of tumor shrinkage to non-irradiated metastatic disease thereby inducing an "abscopal effect." Combined induction of the cGAS-STING pathway and activation of IFN-gamma signaling cascade related to RT within an activated immune environment promotes neoantigen presentation and expansion of cytotoxic effector cells enabling enhancement of systemic immune response. A proposed mechanism, case examples, and clinical trial evidence of "abscopal effect" benefit are reviewed. Results support strategic therapeutic testing to enhance "abscopal effect."
Collapse
Affiliation(s)
- Daniel J. Craig
- University of Toledo, Department of Internal Medicine, Toledo, OH, United States
| | | | - Laura Stanbery
- Medical Affairs, Gradalis, Inc., Carrollton, TX, United States
| | - Adam Walter
- Medical Affairs, Gradalis, Inc., Carrollton, TX, United States
- Gynecologic Oncology, Promedica, Toledo, OH, United States
| | - John Nemunaitis
- Medical Affairs, Gradalis, Inc., Carrollton, TX, United States
| |
Collapse
|
7
|
Trolese MC, Scarpa C, Melfi V, Fabbrizio P, Sironi F, Rossi M, Bendotti C, Nardo G. Boosting the peripheral immune response in the skeletal muscles improved motor function in ALS transgenic mice. Mol Ther 2022; 30:2760-2784. [PMID: 35477657 PMCID: PMC9372324 DOI: 10.1016/j.ymthe.2022.04.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/26/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP1) is one of the most powerful pro-inflammatory chemokines. However, its signalling is pivotal in driving injured axon and muscle regeneration.
Collapse
Affiliation(s)
- Maria Chiara Trolese
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Carlotta Scarpa
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Valentina Melfi
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Paola Fabbrizio
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Francesca Sironi
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Martina Rossi
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;.
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;.
| |
Collapse
|
8
|
Applications of Phyto-Nanotechnology for the Treatment of Neurodegenerative Disorders. MATERIALS 2022; 15:ma15030804. [PMID: 35160749 PMCID: PMC8837051 DOI: 10.3390/ma15030804] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
The strategies involved in the development of therapeutics for neurodegenerative disorders are very complex and challenging due to the existence of the blood-brain barrier (BBB), a closely spaced network of blood vessels and endothelial cells that functions to prevent the entry of unwanted substances in the brain. The emergence and advancement of nanotechnology shows favourable prospects to overcome this phenomenon. Engineered nanoparticles conjugated with drug moieties and imaging agents that have dimensions between 1 and 100 nm could potentially be used to ensure enhanced efficacy, cellular uptake, specific transport, and delivery of specific molecules to the brain, owing to their modified physico-chemical features. The conjugates of nanoparticles and medicinal plants, or their components known as nano phytomedicine, have been gaining significance lately in the development of novel neuro-therapeutics owing to their natural abundance, promising targeted delivery to the brain, and lesser potential to show adverse effects. In the present review, the promising application, and recent trends of combined nanotechnology and phytomedicine for the treatment of neurological disorders (ND) as compared to conventional therapies, have been addressed. Nanotechnology-based efforts performed in bioinformatics for early diagnosis as well as futuristic precision medicine in ND have also been discussed in the context of computational approach.
Collapse
|
9
|
Zhang Y, Fu Y, Zhang C, Jia L, Yao N, Lin Y, Dong Y, Fatima N, Alam N, Wang R, Wang W, Bai L, Zhao S, Liu E. MED1 Deficiency in Macrophages Accelerates Intimal Hyperplasia via ROS Generation and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3010577. [PMID: 34853629 PMCID: PMC8629658 DOI: 10.1155/2021/3010577] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/17/2021] [Accepted: 11/07/2021] [Indexed: 11/17/2022]
Abstract
Mediator complex subunit 1 (MED1) is a component of the mediator complex and functions as a coactivator involved in the regulated transcription of nearly all RNA polymerase II-dependent genes. Previously, we showed that MED1 in macrophages has a protective effect on atherosclerosis; however, the effect of MED1 on intimal hyperplasia and mechanisms regulating proinflammatory cytokine production after macrophage MED1 deletion are still unknown. In this study, we report that MED1 macrophage-specific knockout (MED1 ΔMac) mice showed aggravated neointimal hyperplasia, vascular smooth muscle cells (VSMCs), and macrophage accumulation in injured arteries. Moreover, MED1 ΔMac mice showed increased proinflammatory cytokine production after an injury to the artery. After lipopolysaccharide (LPS) treatment, MED1 ΔMac macrophages showed increased generation of reactive oxygen species (ROS) and reduced expression of peroxisome proliferative activated receptor gamma coactivator-1α (PGC1α) and antioxidant enzymes, including catalase and glutathione reductase. The overexpression of PGC1α attenuated the effects of MED1 deficiency in macrophages. In vitro, conditioned media from MED1 ΔMac macrophages induced more proliferation and migration of VSMCs. To explore the potential mechanisms by which MED1 affects inflammation, macrophages were treated with BAY11-7082 before LPS treatment, and the results showed that MED1 ΔMac macrophages exhibited increased expression of phosphorylated-p65 and phosphorylated signal transducer and activator of transcription 1 (p-STAT1) compared with the control macrophages, suggesting the enhanced activation of NF-κB and STAT1. In summary, these data showed that MED1 deficiency enhanced inflammation and the proliferation and migration of VSMCs in injured vascular tissue, which may result from the activation of NF-κB and STAT1 due to the accumulation of ROS.
Collapse
Affiliation(s)
- Yali Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Yu Fu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Chenyang Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Linying Jia
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Nuo Yao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Yuhao Lin
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Yue Dong
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Nazira Fatima
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Naqash Alam
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Rong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Weirong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Liang Bai
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| |
Collapse
|
10
|
Cognitive and Neuronal Link With Inflammation: A Longitudinal Study in People With and Without HIV Infection. J Acquir Immune Defic Syndr 2021; 85:617-625. [PMID: 32932412 DOI: 10.1097/qai.0000000000002484] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Across many settings, lack of virologic control remains common in people with HIV (PWH) because of late presentation and lack of retention in care. This contributes to neuronal damage and neurocognitive impairment, which remains prevalent. More evidence is needed to understand these outcomes in both PWH and people without HIV (PWOH). METHODS We recruited PWH initiating antiretroviral therapy and PWOH at 2 sites in the United States. One hundred eight adults were enrolled (56 PWOH and 52 PWH), most of whom had a second assessment at least 24 weeks later (193 total assessments). Tumor necrosis factor alpha, monocyte chemotactic protein-1 (MCP-1), neopterin, soluble CD14, and neurofilament light chain protein (NFL) were measured in plasma and cerebrospinal fluid (CSF). Using multivariate models including Bayesian model averaging, we analyzed factors associated with global neuropsychological performance (NPT-9) and CSF NFL at baseline and over time. RESULTS At baseline, higher CSF MCP-1 and plasma sCD14 were associated with worse NPT-9 in PWH, while CSF HIV RNA decrease was the only marker associated with improved NPT-9 over time. Among PWH, higher CSF neopterin was most closely associated with higher NFL. Among PWOH, higher CSF MCP-1 was most closely associated with higher NFL. After antiretroviral therapy initiation, decrease in CSF MCP-1 was most closely associated with NFL decrease. CONCLUSION Monocyte-associated CSF biomarkers are highly associated with neuronal damage in both PWH and PWOH. More research is needed to evaluate whether therapies targeting monocyte-associated inflammation may ameliorate HIV-associated neurobehavioral diseases.
Collapse
|
11
|
Kadry H, Noorani B, Cucullo L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020; 17:69. [PMID: 33208141 PMCID: PMC7672931 DOI: 10.1186/s12987-020-00230-3] [Citation(s) in RCA: 885] [Impact Index Per Article: 177.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/07/2020] [Indexed: 02/07/2023] Open
Abstract
The blood–brain barrier is playing a critical role in controlling the influx and efflux of biological substances essential for the brain’s metabolic activity as well as neuronal function. Thus, the functional and structural integrity of the BBB is pivotal to maintain the homeostasis of the brain microenvironment. The different cells and structures contributing to developing this barrier are summarized along with the different functions that BBB plays at the brain–blood interface. We also explained the role of shear stress in maintaining BBB integrity. Furthermore, we elaborated on the clinical aspects that correlate between BBB disruption and different neurological and pathological conditions. Finally, we discussed several biomarkers that can help to assess the BBB permeability and integrity in-vitro or in-vivo and briefly explain their advantages and disadvantages.
Collapse
Affiliation(s)
- Hossam Kadry
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Luca Cucullo
- Dept. of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Office 415, Rochester, MI, 48309, USA.
| |
Collapse
|
12
|
Alsheikh AJ, Dasinger JH, Abais-Battad JM, Fehrenbach DJ, Yang C, Cowley AW, Mattson DL. CCL2 mediates early renal leukocyte infiltration during salt-sensitive hypertension. Am J Physiol Renal Physiol 2020; 318:F982-F993. [PMID: 32150444 DOI: 10.1152/ajprenal.00521.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Studies examining mechanisms of Dahl salt-sensitive (SS) hypertension have implicated the infiltration of leukocytes in the kidneys, which contribute to renal disease and elevated blood pressure. However, the signaling pathways by which leukocytes traffic to the kidneys remain poorly understood. The present study nominated a signaling pathway by analyzing a kidney RNA sequencing data set from SS rats fed either a low-salt (0.4% NaCl) diet or a high-salt (4.0% NaCl) diet. From this analysis, chemokine (C-C motif) ligand 2 (CCL2) and chemokine (C-C motif) receptor 2 (CCR2) were nominated as a potential pathway modifying renal leukocyte infiltration and contributing to SS hypertension. The functional role of the CCL2/CCR2 pathway was tested by daily administration of CCR2 antagonist (RS-102895 at 5 mg·kg-1·day-1 in DMSO) or DMSO vehicle for 3 or 21 days by intraperitoneal injections during the high salt challenge. Blood pressure, renal leukocyte infiltration, and renal damage were evaluated. The results demonstrated that RS-102895 treatment ameliorated renal damage (urinary albumin excretion; 43.4 ± 5.1 vs. 114.7 ± 15.2 mg/day in vehicle, P < 0.001) and hypertension (144.3 ± 2.2 vs. 158.9 ± 4.8 mmHg in vehicle, P < 0.001) after 21 days of high-salt diet. It was determined that renal leukocyte infiltration was blunted by day 3 of the high-salt diet (1.4 ± 0.1 vs. 1.9 ± 0.2 in vehicle × 106 CD45+ cells/kidney, P = 0.034). An in vitro chemotaxis assay validated the effect of RS-102895 on leukocyte chemotaxis toward CCL2. The results suggest that increased CCL2 in SS kidneys is important in the early recruitment of leukocytes, and blockade of this recruitment by administering RS-102895 subsequently blunted the renal damage and hypertension.
Collapse
Affiliation(s)
- Ammar J Alsheikh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John Henry Dasinger
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Daniel J Fehrenbach
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
13
|
Wang S, Ran Y, Chen X, Li C, Cheng S, Liu J. Pleiotropic Effects of Simvastatin on the Regulation of Potassium Channels in Monocytes. Front Pharmacol 2020; 11:101. [PMID: 32153409 PMCID: PMC7046754 DOI: 10.3389/fphar.2020.00101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 01/28/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose The underlying mechanism of pleiotropic effects of statins on atherosclerosis is still unclear. Kv1.3 and KCa3.1 are two potassium channels that might be involved in monocyte migration and atherosclerosis formation. The aim of this study was to investigate the effect of simvastatin on the Kv1.3 and KCa3.1 in monocyte. Methods and Results In human monocytic THP-1 cells, simvastatin significantly inhibited Kv1.3 mRNA and protein expression by real-time quantitative PCR analysis and western blotting. However, simvastatin had no effects on KCa3.1 mRNA and protein expression. By whole-cell patch clamp, simvastatin (10 μM) remarkably inhibited the current intensity of Kv1.3, but had no effect on KCa3.1. Simvastatin (10 μM) treatment significantly reduced the monocyte chemoattractant protein 1 (MCP-1)-induced monocyte migration. This inhibition was only partially reversed by mevalonate (1mM). In human peripheral blood mononuclear cells (PBMCs), both Kv1.3 and KCa3.1 mRNA expression were increased in patients with coronary artery diseases (CAD) (n = 20) compared to healthy controls (n = 22). However, simvastatin (40 mg per day) significantly inhibited the Kv1.3 but not KCa3.1 mRNA expression after 1 month and 3 months therapy in CAD patients. Conclusion Our data suggested Kv1.3 in monocytes was a potential molecular target of the pleiotropic effects of statins. KCa3.1 might be another marker of CAD, but not associated with statins treatment.
Collapse
Affiliation(s)
- Shaoping Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yuhua Ran
- Department of New Drug Evaluation, State Key Laboratory of Toxicology Medical Courtermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Xuejun Chen
- Research Institute of Chemical Defense, Beijing, China
| | - Chungang Li
- No. 926 Hospital, Joint Logistics Support, Force of PLA, Yunan, China
| | - Shujuan Cheng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Jinghua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
14
|
Gschwandtner M, Derler R, Midwood KS. More Than Just Attractive: How CCL2 Influences Myeloid Cell Behavior Beyond Chemotaxis. Front Immunol 2019; 10:2759. [PMID: 31921102 PMCID: PMC6923224 DOI: 10.3389/fimmu.2019.02759] [Citation(s) in RCA: 434] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1/CCL2) is renowned for its ability to drive the chemotaxis of myeloid and lymphoid cells. It orchestrates the migration of these cell types both during physiological immune defense and in pathological circumstances, such as autoimmune diseases including rheumatoid arthritis and multiple sclerosis, inflammatory diseases including atherosclerosis, as well as infectious diseases, obesity, diabetes, and various types of cancer. However, new data suggest that the scope of CCL2's functions may extend beyond its original characterization as a chemoattractant. Emerging evidence shows that it can impact leukocyte behavior, influencing adhesion, polarization, effector molecule secretion, autophagy, killing, and survival. The direction of these CCL2-induced responses is context dependent and, in some cases, synergistic with other inflammatory stimuli. The involvement of CCL2 signaling in multiple diseases renders it an interesting therapeutic target, although current targeting strategies have not met early expectations in the clinic. A better understanding of how CCL2 affects immune cells will be pivotal to the improvement of existing therapeutic approaches and the development of new drugs. Here, we provide an overview of the pleiotropic effects of CCL2 signaling on cells of the myeloid lineage, beyond chemotaxis, and highlight how these actions might help to shape immune cell behavior and tumor immunity.
Collapse
Affiliation(s)
- Martha Gschwandtner
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Rupert Derler
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Kim S. Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Rizvi SMD, Hussain T, Ahmed ABF, Alshammari TM, Moin A, Ahmed MQ, Barreto GE, Kamal MA, Ashraf GM. Gold nanoparticles: A plausible tool to combat neurological bacterial infections in humans. Biomed Pharmacother 2018; 107:7-18. [PMID: 30075371 DOI: 10.1016/j.biopha.2018.07.130] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/16/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023] Open
Abstract
Management of bacterial infections of central nervous system is a major challenge for the scientists all over the world. Despite the development of various potential drugs, the issue of central nervous system infections persists in the society. The main constraint is the delivery of drugs across the blood brain barrier and only a few drugs after meeting the stringent criteria could cross the blood brain barrier. On the other hand, certain bacterial pathogens could easily enter the brain by using several factors and mechanisms by crossing the blood brain barriers. Interestingly, in the recent past, gold nanoparticles have shown immense potential to overcome the issues associated with the treatment of central nervous system infections, especially due to their inherent ability to cross the blood brain barrier. Initially, the present review summarized the recent updates on the pathogenesis and factors involved in neurological bacterial infections, including the mechanism used by bacterial pathogens to cross the blood brain barriers. Thereafter, the emphasis of the review was on providing current information on gold nanoparticles pertinent to their applicability for the treatment of neurological infections. After discussing the background of neurological bacterial infections, the characteristic features, antibacterial properties, mechanisms of antibacterial action and ability to cross the blood brain barrier of gold nanoparticles have been summarized. Some of the features of gold nanoparticles that make them an ideal candidate for brain delivery are biocompatibity, stability, ability to get synthesized in different sizes with facile methods, surface affinity towards various functional groups, spontaneous crossing of blood brain barrier without applying any external field and most importantly, easy non-invasive tracing by CT imaging. The current updates on the development of gold nanoparticles based therapeutic strategies for the prevention and treatment of central nervous system infections have been discussed in the present study. However, further investigation would be required to translate these preclinical outcomes into clinical applications. Nevertheless, we could safely state that the information gathered and discussed in the present review would benefit the scientists working in the field of neuro-nanotechnology.
Collapse
Affiliation(s)
- Syed Mohd Danish Rizvi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Saudi Arabia.
| | - Talib Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Abo Bakr Fathy Ahmed
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Thamir M Alshammari
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Mohammed Qumani Ahmed
- Department of Pharmacology, College of Medicine,University of Hail, Hail, Saudi Arabia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C, Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Mohammad Ajmal Kamal
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Sydney, Australia; Novel Global Community Educational Foundation, Australia; King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
16
|
Opioids: Modulators of angiogenesis in wound healing and cancer. Oncotarget 2018; 8:25783-25796. [PMID: 28445930 PMCID: PMC5421968 DOI: 10.18632/oncotarget.15419] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/07/2017] [Indexed: 12/12/2022] Open
Abstract
Opioids are potent drugs that are widely used to control wound or cancer pain. Increasing evidence suggest that opioids mediate clinically relevant effects that go beyond their classical role as analgesics. Of note, opioids appear to modulate angiogenesis - a process that is critical in wound healing and cancer progression. In this review, we focus on pro- and anti-angiogenic facets of opioids that arise from the activation of individual opioid receptors and the usage of individual concentrations or application routes. We overview the still incompletely elucidated mechanisms of these angiogenic opioid actions. Moreover, we describe plausible opioids effects, which - although not primarily studied in the context of vessel formation - may be related to the opioid-driven processes of angiogenesis. Finally we discuss the use of opioids as an innovative therapeutic avenue for the treatment of chronic wounds and cancer.
Collapse
|
17
|
Leibrand CR, Paris JJ, Ghandour MS, Knapp PE, Kim WK, Hauser KF, McRae M. HIV-1 Tat disrupts blood-brain barrier integrity and increases phagocytic perivascular macrophages and microglia in the dorsal striatum of transgenic mice. Neurosci Lett 2017; 640:136-143. [PMID: 28057474 DOI: 10.1016/j.neulet.2016.12.073] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/19/2016] [Accepted: 12/31/2016] [Indexed: 12/01/2022]
Abstract
HIV-1 infection results in blood-brain barrier (BBB) disruption, which acts as a rate-limiting step for HIV-1 entry into the CNS and for subsequent neuroinflammatory/neurotoxic actions. One mechanism by which HIV may destabilize the BBB involves actions of the HIV-1 regulatory protein, trans-activator of transcription (Tat). We utilized a conditional, Tat-expressing transgenic murine model to examine the influence of Tat1-86 expression on BBB integrity and to assess the relative numbers of phagocytic perivascular macrophages and microglia within the CNS in vivo. The effects of Tat exposure on sodium-fluorescein (Na-F; 0.376kDa), horseradish peroxidase (HRP; 44kDa), and Texas Red-labeled dextran (70kDa) leakage into the brain were assessed in Tat-exposed (Tat+) and control (Tat-) mice. Exposure to HIV-1 Tat significantly increased both Na-F and HRP, but not the larger sized Texas Red-labeled dextran, confirming BBB breakdown and also suggesting the breach was limited to molecules <70kDa. Additionally, at 5 d after Tat induction, Alexa Fluor® 488-labeled dextran was bilaterally infused into the lateral ventricles 5 d before the termination of the experiment. Within the caudate/putamen, Tat induction increased the proportion of dextran-labeled Iba-1+ phagocytic perivascular macrophages (∼5-fold) and microglia (∼3-fold) compared to Tat- mice. These data suggest that HIV-1 Tat exposure is sufficient to destabilize BBB integrity and to increase the presence of activated, phagocytic, perivascular macrophages and microglia in an in vivo model of neuroAIDS.
Collapse
Affiliation(s)
- Crystal R Leibrand
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jason J Paris
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, P.O. Box 980613, Richmond, VA 23298, USA
| | - M Said Ghandour
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, P.O. Box 980709, Richmond, VA 23298, USA
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, P.O. Box 980613, Richmond, VA 23298, USA; Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, P.O. Box 980709, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298, USA
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, P.O. Box 980613, Richmond, VA 23298, USA; Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, P.O. Box 980709, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
18
|
Sela H, Cohen H, Elia P, Zach R, Karpas Z, Zeiri Y. Spontaneous penetration of gold nanoparticles through the blood brain barrier (BBB). J Nanobiotechnology 2015; 13:71. [PMID: 26489846 PMCID: PMC4618365 DOI: 10.1186/s12951-015-0133-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/06/2015] [Indexed: 01/17/2023] Open
Abstract
Background The blood brain barrier (BBB) controls the brain microenvironment and limits penetration of the central nervous system (CNS) by chemicals, thus creating an obstacle to many medical imaging and treatment procedures. Research efforts to identify viable routes of BBB penetration have focused on structures such as micelles, polymeric nanoparticles and liposomes as drug carriers, however, many of them failed to provide unequivocal proof of BBB penetration. Here we proved that gold nanoparticles (AuNPs) penetrate the BBB in rats to reach brain regions. Results Injection of AuNPs to the abdominal cavity of rats resulted in levels of gold found in blood, urine, brain regions and body organs. After perfusion the concentration of gold in brain regions diminished dramatically indicating that most of the gold was in venous blood and not in the brain tissues. Injection of Na, K or Ca ion channel blockers reduced BBB penetration by half. A biological half-life of 12.9 ± 4.9 h was found for the gold nanoparticles. Possible mechanisms for the transport of AuNPs through the BBB are discussed. Conclusions BBB penetration by AuNPs is spontaneous without the application of an external field. A major amount of gold resides in blood vessels therefore perfusion required. Ion channel blockers can be used to control the transport of AuNPs. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0133-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hagit Sela
- Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel. .,Department of Chemistry, NRCN, P.O. Box 9001, Beer-Sheva, 8419001, Israel.
| | - Hagit Cohen
- The State of Israel Ministry of Health, Anxiety and Stress Research Unit, Faculty of Health Sciences, Beer-Sheva Mental Health Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Paz Elia
- Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
| | - Raya Zach
- Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
| | - Zeev Karpas
- Department of Chemistry, NRCN, P.O. Box 9001, Beer-Sheva, 8419001, Israel.
| | - Yehuda Zeiri
- Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel. .,Department of Chemistry, NRCN, P.O. Box 9001, Beer-Sheva, 8419001, Israel.
| |
Collapse
|
19
|
Haley SA, O'Hara BA, Nelson CDS, Brittingham FLP, Henriksen KJ, Stopa EG, Atwood WJ. Human polyomavirus receptor distribution in brain parenchyma contrasts with receptor distribution in kidney and choroid plexus. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2246-58. [PMID: 26056932 DOI: 10.1016/j.ajpath.2015.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/21/2015] [Accepted: 04/14/2015] [Indexed: 11/25/2022]
Abstract
The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy, a rare demyelinating disease that occurs in the setting of prolonged immunosuppression. After initial asymptomatic infection, the virus establishes lifelong persistence in the kidney and possibly other extraneural sites. In rare instances, the virus traffics to the central nervous system, where oligodendrocytes, astrocytes, and glial precursors are susceptible to lytic infection, resulting in progressive multifocal leukoencephalopathy. The mechanisms by which the virus traffics to the central nervous system from peripheral sites remain unknown. Lactoseries tetrasaccharide c (LSTc), a pentasaccharide containing a terminal α2,6-linked sialic acid, is the major attachment receptor for polyomavirus. In addition to LSTc, type 2 serotonin receptors are required for facilitating virus entry into susceptible cells. We studied the distribution of virus receptors in kidney and brain using lectins, antibodies, and labeled virus. The distribution of LSTc, serotonin receptors, and virus binding sites overlapped in kidney and in the choroid plexus. In brain parenchyma, serotonin receptors were expressed on oligodendrocytes and astrocytes, but these cells were negative for LSTc and did not bind virus. LSTc was instead found on microglia and vascular endothelium, to which virus bound abundantly. Receptor distribution was not changed in the brains of patients with progressive multifocal leukoencephalopathy. Virus infection of oligodendrocytes and astrocytes during disease progression is LSTc independent.
Collapse
Affiliation(s)
- Sheila A Haley
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Bethany A O'Hara
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Christian D S Nelson
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Frances L P Brittingham
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Kammi J Henriksen
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Edward G Stopa
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Walter J Atwood
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island.
| |
Collapse
|
20
|
Chen D, Yu SP, Wei L. Ion channels in regulation of neuronal regenerative activities. Transl Stroke Res 2014; 5:156-62. [PMID: 24399572 DOI: 10.1007/s12975-013-0320-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/18/2013] [Accepted: 12/20/2013] [Indexed: 02/08/2023]
Abstract
The regeneration of the nervous system is achieved by the regrowth of damaged neuronal axons, the restoration of damaged nerve cells, and the generation of new neurons to replace those that have been lost. In the central nervous system, the regenerative ability is limited by various factors including damaged oligodendrocytes that are essential for neuronal axon myelination, an emerging glial scar, and secondary injury in the surrounding areas. Stem cell transplantation therapy has been shown to be a promising approach to treat neurodegenerative diseases because of the regenerative capability of the stem cells that secrete neurotrophic factors and give rise to differentiated progeny. However, some issues of stem cell transplantation, such as survival, homing, and efficiency of neural differentiation after transplantation, still need to be improved. Ion channels allow for the exchange of ions between the intra- and extracellular spaces or between the cytoplasm and organelles. These ion channels maintain the ion homeostasis in the brain and play a key role in regulating the physiological function of the nervous system and allowing the processing of neuronal signals. In seeking a potential strategy to enhance the efficacy of stem cell therapy in neurological and neurodegenerative diseases, this review briefly summarizes the roles of ion channels in cell proliferation, differentiation, migration, chemotropic axon guidance of growth cones, and axon outgrowth after injury.
Collapse
Affiliation(s)
- Dongdong Chen
- Department of Anesthesiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
21
|
Tatro ET, Hefler S, Shumaker-Armstrong S, Soontornniyomkij B, Yang M, Yermanos A, Wren N, Moore DJ, Achim CL. Modulation of BK channel by MicroRNA-9 in neurons after exposure to HIV and methamphetamine. J Neuroimmune Pharmacol 2013; 8:1210-23. [PMID: 23508624 PMCID: PMC3715589 DOI: 10.1007/s11481-013-9446-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/26/2013] [Indexed: 10/27/2022]
Abstract
MicroRNAs (miR) regulate phenotype and function of neurons by binding to miR-response elements (MRE) in the 3' untranslated regions (3'UTR) of various messenger RNAs to inhibit translation. MiR expression can be induced or inhibited by environmental factors like drug exposure and viral infection, leading to changes in cellular physiology. We hypothesized that the effects of methamphetamine (MA) and human immunodeficiency virus (HIV)-infection in the brain will induce changes in miR expression, and have downstream regulatory consequences in neurons. We first used a PCR-based array to screen for differential expression of 380 miRs in frontal cortex autopsy tissues of HIV-positive MA abusers and matched controls. These results showed significantly increased expression of the neuron-specific miR-9. In vitro, we used SH-SY5Y cells, an experimental system for dopaminergic studies, to determine miR expression by quantitative PCR after exposure to MA in the presence or absence of conditioned media from HIV-infected macrophages. Again, we found that miR-9 was significantly increased compared to controls. We also examined the inwardly rectifying potassium channel, KCNMA1, which has alternative splice variants that contain an MRE to miR-9. We identified alternate 3'UTRs of KCNMA1 both in vitro and in the autopsy specimens and found differential splice variant expression of KCNMA1, operating via the increased miR-9. Our results suggest that HIV and MA -induced elevated miR-9, leading to suppression of MRE-containing splice variants of KCNMA1, which may affect neurotransmitter release in dopaminergic neurons.
Collapse
Affiliation(s)
- Erick T Tatro
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chang YN, Guo H, Li J, Song Y, Zhang M, Jin J, Xing G, Zhao Y. Adjusting the balance between effective loading and vector migration of macrophage vehicles to deliver nanoparticles. PLoS One 2013; 8:e76024. [PMID: 24116086 PMCID: PMC3792996 DOI: 10.1371/journal.pone.0076024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/22/2013] [Indexed: 11/18/2022] Open
Abstract
The nature of macrophage allows the possibility that this cell type could be used as drug delivery system to track therapeutic drug nanoparticles (NPs) in cancer. However, there is no existing research on the regulation between effective loading of NPs and targeted delivery of macrophages. Here, we investigated the important parameters of intracellular NP quantity and the vector migration rate. Macrophage loading capacity was obtained by comparing the uptake quantity of varisized NPs, and the delivery ability of loaded cells was determined by measuring vector migration rates. We observed a positive correlation between the size of NPs and directed macrophage migration. Our findings suggest that the molecular mechanism of migration vector rate regulation involved increased expression levels of colony-stimulating factor-1 (CSF-1) receptor and integrin induced by 100-nm and 500-nm particles. The ability of macrophages uptake to varisized NPs showed the opposite trend, with the increased vector rate of cell migration influenced by NPs. We are able to demonstrate the important balance between effective macrophage loading and targeted delivery. By adjusting the balance parameters, it will be possible to utilize NPs in macrophage-mediated disease diagnosis and therapy.
Collapse
Affiliation(s)
- Ya-Nan Chang
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Haili Guo
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Juan Li
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Yan Song
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Mingyi Zhang
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Junjiang Jin
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Gengmei Xing
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
- * E-mail: (GX); (YZ)
| | - Yuliang Zhao
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
- Chinese Academy of Science Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, China
- * E-mail: (GX); (YZ)
| |
Collapse
|
23
|
Anti-inflammatory and carbonic anhydrase restoring actions of yam powder (Dioscorea spp) contribute to the prevention of cysteamine-induced duodenal ulcer in a rat model. Nutr Res 2013; 33:677-85. [PMID: 23890358 DOI: 10.1016/j.nutres.2013.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 05/29/2013] [Accepted: 05/31/2013] [Indexed: 01/11/2023]
Abstract
Increased acid output, accompanied with a defective defense system, is considered a fundamental pathogenesis of duodenal ulcer (DU). However, relapse of DU occurs despite proton pump inhibitors and H2 receptor antagonists, hence imposing the enforcement of the defense system. Dried powder of the yam tuber (Dioscorea spp) has been used in traditional folk medicine as a nutritional fortification. We hypothesized that dried-yam powder would prevent DU through improvement of anti-inflammatory actions and carbonic anhydrase (CA) activity. Therefore, we investigated the preventive effects of dried-yam powder against the cysteamine-induced DU and elucidated the underlying mechanisms. Duodenal ulcers were induced in Sprague-Dawley rats by intragastric administration of 500 mg/kg cysteamine-HCl. The dried-yam powder was used as a pretreatment before the cysteamine-HCl. The number and size of DU were measured. The expressions of inflammation mediators were checked in duodenal tissues, and the expressions of CAs and malondialdehyde levels were also examined. Cysteamine provoked perforated DU, whereas dried-yam powder significantly prevented DU as much as pantoprazole and significantly reduced the incidence of perforation. The messenger RNA expressions of cyclooxygenase-2 and inducible nitric oxide synthase were remarkably decreased in the yam group compared with the cysteamine group, and the serum levels of proinflammatory cytokines including interleukin-1β, interleukin-6, and tumor necrosis factor were significantly attenuated in the yam group. Cysteamine significantly decreased the expression of CAs, whereas yam treatment significantly preserved the expressions of CA IX, XII, and XIV. In conclusion, dried-yam powder exerts a significant protective effect against cysteamine-induced DU by lowering the activity of inflammatory cytokines and free radicals and restoring the activity of CAs, except in CA IV.
Collapse
|
24
|
Dohgu S, Banks WA. Brain pericytes increase the lipopolysaccharide-enhanced transcytosis of HIV-1 free virus across the in vitro blood-brain barrier: evidence for cytokine-mediated pericyte-endothelial cell crosstalk. Fluids Barriers CNS 2013; 10:23. [PMID: 23816186 PMCID: PMC3710206 DOI: 10.1186/2045-8118-10-23] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 06/23/2013] [Indexed: 11/30/2024] Open
Abstract
Background Human immunodeficiency virus-1 (HIV-1) enters the brain by crossing the blood–brain barrier (BBB) as both free virus and within infected immune cells. Previous work showed that activation of the innate immune system with lipopolysaccharide (LPS) enhances free virus transport both in vivo and across monolayer monocultures of brain microvascular endothelial cells (BMECs) in vitro. Methods Here, we used monocultures and co-cultures of brain pericytes and brain endothelial cells to examine the crosstalk between these cell types in mediating the LPS-enhanced permeation of radioactively-labeled HIV-1 (I-HIV) across BMEC monolayers. Results We found that brain pericytes when co-cultured with BMEC monolayers magnified the LPS-enhanced transport of I-HIV without altering transendothelial electrical resistance, indicating that pericytes affected the transcytotic component of HIV-1 permeation. As LPS crosses the BBB poorly if at all, and since pericytes are on the abluminal side of the BBB, we postulated that luminal LPS acts indirectly on pericytes through abluminal secretions from BMECs. Consistent with this, we found that the pattern of secretion of cytokines by pericytes directly exposed to LPS was different than when the pericytes were exposed to the abluminal fluid from LPS-treated BMEC monolayers. Conclusion These results are evidence for a cellular crosstalk in which LPS acts at the luminal surface of the brain endothelial cell, inducing abluminal secretions that stimulate pericytes to release substances that enhance the permeability of the BMEC monolayer to HIV.
Collapse
Affiliation(s)
- Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| | | |
Collapse
|
25
|
Wang T, Xu Y, Zhu H, Andrus T, Ivanov SB, Pan C, Dolores J, Dann GC, Zhou M, Forte D, Yang Z, Holte S, Corey L, Zhu T. Successful isolation of infectious and high titer human monocyte-derived HIV-1 from two subjects with discontinued therapy. PLoS One 2013; 8:e65071. [PMID: 23741458 PMCID: PMC3669022 DOI: 10.1371/journal.pone.0065071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 04/20/2013] [Indexed: 12/13/2022] Open
Abstract
Background HIV-1 DNA in blood monocytes is considered a viral source of various HIV-1 infected tissue macrophages, which is also known as “Trojan horse” hypothesis. However, whether these DNA can produce virions has been an open question for years, due to the inability of isolating high titer and infectious HIV-1 directly from monocytes. Results In this study, we demonstrated successful isolation of two strains of M-HIV-1 (1690 M and 1175 M) from two out of four study subjects, together with their in vivo controls, HIV-1 isolated from CD4+ T-cells (T-HIV-1), 1690 T and 1175 T. All M- and T- HIV-1 isolates were detected CCR5-tropic. Both M- HIV-1 exhibited higher levels of replication in monocyte-derived macrophages (MDM) than the two T- HIV-1. Consistent with our previous reports on the subject 1175 with late infection, compartmentalized env C2-V3-C3 sequences were identified between 1175 M and 1175 T. In contrast, 1690 M and 1690 T, which were isolated from subject 1690 with relatively earlier infection, showed homogenous env C2-V3-C3 sequences. However, multiple reverse transcriptase (RT) inhibitor resistance-associated variations were detected in the Gag-Pol region of 1690 M, but not of 1690 T. By further measuring HIV DNA intracellular copy numbers post-MDM infection, 1690 M was found to have significantly higher DNA synthesis efficiency than 1690 T in macrophages, indicating a higher RT activity, which was confirmed by AZT inhibitory assays. Conclusions These results suggested that the M- and T- HIV-1 are compartmentalized in the two study subjects, respectively. Therefore, we demonstrated that under in vitro conditions, HIV-1 infected human monocytes can productively release live viruses while differentiating into macrophages.
Collapse
Affiliation(s)
- Tong Wang
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Guangzhou, Guangdong, China
- Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Younong Xu
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Haiying Zhu
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Thomas Andrus
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sergei B. Ivanov
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Charlotte Pan
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jazel Dolores
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Gregory C. Dann
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Michael Zhou
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Dominic Forte
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Zihuan Yang
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sarah Holte
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lawrence Corey
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Division of Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Tuofu Zhu
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Division of Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
26
|
Liu J, Xu P, Collins C, Liu H, Zhang J, Keblesh JP, Xiong H. HIV-1 Tat protein increases microglial outward K(+) current and resultant neurotoxic activity. PLoS One 2013; 8:e64904. [PMID: 23738010 PMCID: PMC3667810 DOI: 10.1371/journal.pone.0064904] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 04/19/2013] [Indexed: 11/19/2022] Open
Abstract
Microglia plays a crucial role in the pathogenesis of HIV-1-associated neurocognitive disorders. Increasing evidence indicates the voltage-gated potassium (Kv) channels are involved in the regulation of microglia function, prompting us to hypothesize Kv channels may also be involved in microglia-mediated neurotoxic activity in HIV-1-infected brain. To test this hypothesis, we investigated the involvement of Kv channels in the response of microglia to HIV-1 Tat protein. Treatment of rat microglia with HIV-1 Tat protein (200 ng/ml) resulted in pro-inflammatory microglial activation, as indicated by increases in TNF-α, IL-1β, reactive oxygen species, and nitric oxide, which were accompanied by enhanced outward K(+) current and Kv1.3 channel expression. Suppression of microglial Kv1.3 channel activity, either with Kv1.3 channel blockers Margatoxin, 5-(4-Phenoxybutoxy)psoralen, or broad-spectrum K(+) channel blocker 4-Aminopyridine, or by knockdown of Kv1.3 expression via transfection of microglia with Kv1.3 siRNA, was found to abrogate the neurotoxic activity of microglia resulting from HIV-1 Tat exposure. Furthermore, HIV-1 Tat-induced neuronal apoptosis was attenuated with the application of supernatant collected from K(+) channel blocker-treated microglia. Lastly, the intracellular signaling pathways associated with Kv1.3 were investigated and enhancement of microglial Kv1.3 was found to correspond with an increase in Erk1/2 mitogen-activated protein kinase activation. These data suggest targeting microglial Kv1.3 channels may be a potential new avenue of therapy for inflammation-mediated neurological disorders.
Collapse
Affiliation(s)
- Jianuo Liu
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (JL); (HX)
| | - Peng Xu
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Cory Collins
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Han Liu
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jingdong Zhang
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - James P. Keblesh
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Huangui Xiong
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (JL); (HX)
| |
Collapse
|
27
|
Schwab A, Fabian A, Hanley PJ, Stock C. Role of ion channels and transporters in cell migration. Physiol Rev 2013; 92:1865-913. [PMID: 23073633 DOI: 10.1152/physrev.00018.2011] [Citation(s) in RCA: 328] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell motility is central to tissue homeostasis in health and disease, and there is hardly any cell in the body that is not motile at a given point in its life cycle. Important physiological processes intimately related to the ability of the respective cells to migrate include embryogenesis, immune defense, angiogenesis, and wound healing. On the other side, migration is associated with life-threatening pathologies such as tumor metastases and atherosclerosis. Research from the last ≈ 15 years revealed that ion channels and transporters are indispensable components of the cellular migration apparatus. After presenting general principles by which transport proteins affect cell migration, we will discuss systematically the role of channels and transporters involved in cell migration.
Collapse
|
28
|
Liu J, Xu C, Chen L, Xu P, Xiong H. Involvement of Kv1.3 and p38 MAPK signaling in HIV-1 glycoprotein 120-induced microglia neurotoxicity. Cell Death Dis 2012; 3:e254. [PMID: 22258405 PMCID: PMC3270274 DOI: 10.1038/cddis.2011.140] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Inflammatory responses mediated by activated microglia play a pivotal role in the pathogenesis of human immunodeficiency virus type 1 (HIV-1)-associated neurocognitive disorders. Studies on identification of specific targets to control microglia activation and resultant neurotoxic activity are imperative. Increasing evidence indicate that voltage-gated K+ (Kv) channels are involved in the regulation of microglia functionality. In this study, we investigated Kv1.3 channels in the regulation of neurotoxic activity mediated by HIV-1 glycoprotein 120 (gp120)-stimulated rat microglia. Our results showed treatment of microglia with gp120 increased the expression levels of Kv1.3 mRNA and protein. In parallel, whole-cell patch-clamp studies revealed that gp120 enhanced microglia Kv1.3 current, which was blocked by margatoxin, a Kv1.3 blocker. The association of gp120 enhancement of Kv1.3 current with microglia neurotoxicity was demonstrated by experimental results that blocking microglia Kv1.3 attenuated gp120-associated microglia production of neurotoxins and neurotoxicity. Knockdown of Kv1.3 gene by transfection of microglia with Kv1.3-siRNA abrogated gp120-associated microglia neurotoxic activity. Further investigation unraveled an involvement of p38 MAPK in gp120 enhancement of microglia Kv1.3 expression and resultant neurotoxic activity. These results suggest not only a role Kv1.3 may have in gp120-associated microglia neurotoxic activity, but also a potential target for the development of therapeutic strategies.
Collapse
Affiliation(s)
- J Liu
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | | | |
Collapse
|
29
|
Strazza M, Pirrone V, Wigdahl B, Nonnemacher MR. Breaking down the barrier: the effects of HIV-1 on the blood-brain barrier. Brain Res 2011; 1399:96-115. [PMID: 21641584 DOI: 10.1016/j.brainres.2011.05.015] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 05/06/2011] [Accepted: 05/07/2011] [Indexed: 01/13/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) primarily infects CD4(+) T cells and cells of the monocyte-macrophage lineage, resulting in immunodeficiency in an infected patient. Along with this immune deficiency, HIV-1 has been linked to a number of neurological symptoms in the absence of opportunistic infections or other co-morbidities, suggesting that HIV-1 is able to cross the blood-brain barrier (BBB), enter the central nervous system (CNS), and cause neurocognitive impairment. HIV-1-infected monocyte-macrophages traverse the BBB and enter the CNS throughout the course of HIV-1 disease. Once in the brain, both free virus and virus-infected cells are able to infect neighboring resident microglia and astrocytes and possibly other cell types. HIV-1-infected cells in both the periphery and the CNS give rise to elevated levels of viral proteins, including gp120, Tat, and Nef, and of host inflammatory mediators such as cytokines and chemokines. It has been shown that the viral proteins may act alone or in concert with host cytokines and chemokines, affecting the integrity of the BBB. The pathological end point of these interactions may facilitate a positive feedback loop resulting in increased penetration of HIV into the CNS. It is proposed in this review that the dysregulation of the BBB during and after neuroinvasion is a critical component of the neuropathogenic process and that dysregulation of this protective barrier is caused by a combination of viral and host factors including secreted viral proteins, components of the inflammatory process, the aging process, therapeutics, and drug or alcohol abuse.
Collapse
Affiliation(s)
- Marianne Strazza
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | | | | | | |
Collapse
|
30
|
Cucullo L, Hossain M, Puvenna V, Marchi N, Janigro D. The role of shear stress in Blood-Brain Barrier endothelial physiology. BMC Neurosci 2011; 12:40. [PMID: 21569296 PMCID: PMC3103473 DOI: 10.1186/1471-2202-12-40] [Citation(s) in RCA: 312] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 05/11/2011] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND One of the most important and often neglected physiological stimuli contributing to the differentiation of vascular endothelial cells (ECs) into a blood-brain barrier (BBB) phenotype is shear stress (SS). With the use of a well established humanized dynamic in vitro BBB model and cDNA microarrays, we have profiled the effect of SS in the induction/suppression of ECs genes and related functions. RESULTS Specifically, we found a significant upregulation of tight and adherens junctions proteins and genes. Trans-endothelial electrical resistance (TEER) and permeability measurements to know substances have shown that SS promoted the formation of a tight and highly selective BBB. SS also increased the RNA level of multidrug resistance transporters, ion channels, and several p450 enzymes. The RNA level of a number of specialized carrier-mediated transport systems (e.g., glucose, monocarboxylic acid, etc.) was also upregulated.RNA levels of modulatory enzymes of the glycolytic pathway (e.g., lactate dehydrogenase) were downregulated by SS while those involved in the Krebs cycle (e.g., lactate and other dehydrogenases) were upregulated. Measurements of glucose consumption versus lactate production showed that SS negatively modulated the glycolytic bioenergetic pathways of glucose metabolism in favor of the more efficient aerobic respiration. BBB ECs are responsive to inflammatory stimuli. Our data showed that SS increased the RNA levels of integrins and vascular adhesion molecules. SS also inhibited endothelial cell cycle via regulation of BTG family proteins encoding genes. This was paralleled by significant increase in the cytoskeletal protein content while that of membrane, cytosol, and nuclear sub-cellular fractions decreased. Furthermore, analysis of 2D gel electrophoresis (which allows identifying a large number of proteins per sample) of EC proteins extracted from membrane sub-cellular endothelial fractions showed that SS increased the expression levels of tight junction proteins. In addition, regulatory enzymes of the Krebb's cycle (aerobic glucose metabolism) were also upregulated. Furthermore, the expression pattern of key protein regulators of the cell cycle and parallel gene array data supported a cell proliferation inhibitory role for SS. CONCLUSIONS Genomic and proteomic analyses are currently used to examine BBB function in healthy and diseased brain and characterize this dynamic interface. In this study we showed that SS plays a key role in promoting the differentiation of vascular endothelial cells into a truly BBB phenotype. SS affected multiple aspect of the endothelial physiology spanning from tight junctions formation to cell division as well as the expression of multidrug resistance transporters. BBB dysfunction has been observed in many neurological diseases, but the causes are generally unknown. Our study provides essential insights to understand the role played by SS in the BBB formation and maintenance.
Collapse
Affiliation(s)
- Luca Cucullo
- Cerebrovascular Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195 USA.
| | | | | | | | | |
Collapse
|
31
|
Relationships between markers of vascular dysfunction and neurodevelopmental outcomes in perinatally HIV-infected youth. AIDS 2010; 24:1481-91. [PMID: 20539091 DOI: 10.1097/qad.0b013e32833a241b] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the relationship between markers of vascular dysfunction and neurodevelopmental status in pediatric HIV disease. DESIGN A cross-sectional design within a prospective, 15-site cohort study conducted in the United States. METHODS Nine vascular biomarkers were examined in 89 HIV-infected children: soluble P-selectin/sCD62P, fibrinogen, adiponectin, monocyte chemoattractant protein-1/CCL-2, interleukin-6, C-reactive protein, soluble vascular cell adhesion molecule-1/sCD106, sE-selectin/sCD62E, and soluble intercellular adhesion molecule-1/sCD54. The Wechsler Intelligence Scale for Children-Fourth edition (WISC-IV) was administered yielding indices for verbal comprehension, perceptual reasoning, working memory and processing speed, and overall composite Full-Scale IQ score. Linear regression models were used to evaluate neurodevelopmental status (measured by WISC-IV scores) as a function of each biomarker while adjusting for demographics, disease severity, and receipt of HAART. Biomarker levels were evaluated in quartiles to evaluate trends in WISC-IV responses. RESULTS Among the 89 HIV-infected children (median age = 12 years), 56% were girls, 71% black, 16% Hispanic, and 43% had yearly household income below US $20,000. Log (soluble P-selectin) was significantly correlated with all WISC-IV scores; adjusted slopes showed 6-11-point average decrease in scores for each one log unit increase in soluble P-selectin. Final linear regression models for log (fibrinogen) adjusted for sociodemographic and disease characteristics also indicated a negative correlation with all WISC-IV scores (13-30-point decrease for each one log unit increase in fibrinogen); these decreases were significant in the verbal comprehension, perceptual reasoning, and Full-Scale IQ scores. CONCLUSION Proinflammatory microvascular and immunologic mechanisms may be involved in neurodevelopmental impairment in children with perinatally acquired HIV disease.
Collapse
|
32
|
Gras G, Kaul M. Molecular mechanisms of neuroinvasion by monocytes-macrophages in HIV-1 infection. Retrovirology 2010; 7:30. [PMID: 20374632 PMCID: PMC2864195 DOI: 10.1186/1742-4690-7-30] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 04/07/2010] [Indexed: 12/12/2022] Open
Abstract
HIV associated neurocognitive disorders and their histopathological correlates largely depend on the continuous seeding of the central nervous system with immune activated leukocytes, mainly monocytes/macrophages from the periphery. The blood-brain-barrier plays a critical role in this never stopping neuroinvasion, although it appears unaltered until the late stage of HIV encephalitis. HIV flux that moves toward the brain thus relies on hijacking and exacerbating the physiological mechanisms that govern blood brain barrier crossing rather than barrier disruption. This review will summarize the recent data describing neuroinvasion by HIV with a focus on the molecular mechanisms involved.
Collapse
Affiliation(s)
- Gabriel Gras
- Institute of Emerging Diseases and Innovative Therapies, Division of Immuno-Virology, CEA, 18 Route du Panorama, F92265 Fontenay-aux Roses, France.
| | | |
Collapse
|
33
|
Das S, Ghosh D, Basu A. Japanese encephalitis virus induce immuno-competency in neural stem/progenitor cells. PLoS One 2009; 4:e8134. [PMID: 19956550 PMCID: PMC2780913 DOI: 10.1371/journal.pone.0008134] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 11/09/2009] [Indexed: 01/19/2023] Open
Abstract
Background The low immunogenicity of neural stem/progenitor cells (NSPCs) coupled with negligible expression of MHC antigens has popularized their use in transplantation medicine. However, in an inflammatory environment, the NSPCs express costimulatory molecules and MHC antigens, and also exhibit certain immunomodulatory functions. Since NSPCs are the cellular targets in a number of virus infections both during postnatal and adult stages, we wanted to investigate the immunological properties of these stem cells in response to viral pathogen. Methodology/Principal Findings We utilized both in vivo mouse model and in vitro neurosphere model of Japanese encephalitis virus (JEV) infection for the study. The NSPCs residing in the subventricular zone of the infected brains showed prominent expression of MHC-I and costimulatory molecules CD40, CD80, and CD86. Using Flow cytometry and fluorescence microscopy, we observed increased surface expression of co-stimulatory molecule and MHC class I antigen in NSPCs upon progressive JEV infection in vitro. Moreover, significant production of pro-inflammatory cyto/chemokines was detected in JEV infected NSPCs by Cytokine Bead Array analysis. Interestingly, NSPCs were capable of providing functional costimulation to allogenic T cells and JEV infection resulted in increased proliferation of allogenic T cells, as detected by Mixed Lymphocyte reaction and CFSE experiments. We also report IL-2 production by NSPCs upon JEV infection, which possibly provides mitogenic signals to T cells and trigger their proliferation. Conclusion/Significance The in vivo and in vitro findings clearly indicate the development of immunogenicity in NSPCs following progressive JEV infection, in our case, JEV infection. Following a neurotropic virus infection, NSPCs possibly behave as immunogenic cells and contribute to both the innate and adaptive immune axes. The newly discovered immunological properties of NSPCs may have implications in assigning a new role of these cells as non-professional antigen presenting cells in the central nervous system.
Collapse
Affiliation(s)
- Sulagna Das
- National Brain Research Centre, Manesar, Haryana, India
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, India
- * E-mail:
| |
Collapse
|
34
|
Abstract
A broad range of nanomedicines is being developed to improve drug delivery for CNS disorders. The structure of the blood-brain barrier (BBB), the presence of efflux pumps and the expression of metabolic enzymes pose hurdles for drug-brain entry. Nanoformulations can circumvent the BBB to improve CNS-directed drug delivery by affecting such pumps and enzymes. Alternatively, they can be optimized to affect their size, shape, and protein and lipid coatings to facilitate drug uptake, release and ingress across the barrier. This is important as the brain is a sanctuary for a broad range of pathogens including HIV-1. Improved drug delivery to the CNS would affect pharmacokinetic and drug biodistribution properties. This article focuses on how nanotechnology can serve to improve the delivery of antiretroviral medicines, termed nanoART, across the BBB and affect the biodistribution and clinical benefit for HIV-1 disease.
Collapse
Affiliation(s)
- Ari Nowacek
- Department of Pharmacology & Experimental Neuroscience, Center for Neurovirology & Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | |
Collapse
|
35
|
Sun J, Mei Y, Guo X, Yin X, Zhao X, Wang Z, Yang L. Voltage-dependent K+-channel responses during activation and damage in alveolar macrophages induced by quartz particles. ACTA ACUST UNITED AC 2009; 29:404-8. [DOI: 10.1007/s11596-009-0402-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Indexed: 10/19/2022]
|
36
|
Ivey NS, MacLean AG, Lackner AA. Acquired immunodeficiency syndrome and the blood-brain barrier. J Neurovirol 2009; 15:111-22. [PMID: 19306229 DOI: 10.1080/13550280902769764] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) plays a critical role in normal physiology of the central nervous system by regulating what reaches the brain from the periphery. The BBB also plays a major role in neurologic disease including neuropathologic sequelae associated with infection by human immunodeficiency virus (HIV) in humans and the closely related simian immunodeficiency virus (SIV) in macaques. In this review, we provide an overview of the function, structure, and components of the BBB, followed by a more detailed discussion of the subcellular structures and regulation of the tight junction. We then discuss the ways in which HIV/SIV affects the BBB, largely through infection of monocytes/macrophages, and how infected macrophages crossing the BBB ultimately results in breakdown of the barrier.
Collapse
Affiliation(s)
- Nathan S Ivey
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | | | | |
Collapse
|
37
|
Nanobiology for the pharmacology of cellular ion channels. J Neuroimmune Pharmacol 2009; 4:7-9. [PMID: 19169828 DOI: 10.1007/s11481-008-9144-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 12/17/2008] [Indexed: 10/21/2022]
Abstract
Writing this editorial is especially pleasing. First, it provides us an opportunity to introduce new directives to the field of Neuroimmune Pharmacology and to explain why the field of nanomedicine is likely an important part of its future growth and development. Second, it is an opportunity to showcase research in this area currently operative in Russia that may not be readily accessible to the readership. Third, it is a platform to better explain why the Journal Editorial leadership was enthusiastic about the science and its relationship to the Society on NeuroImmune Pharmacology strategic goals. All are brought to bear in this issue of the Journal of Neuroimmune Pharmacology. The issue includes articles presented at a recent joint US-Russian workshop entitled, "Health in the 21st Century: Nanomedicine and Self-Organization of Biological Systems" held at M.V. Lomonosov Moscow State University (MSU), Moscow, Russia, December 10-11, 2007. The conjoint meeting was organized through the Departments of Biology, Chemistry, and Physics, MSU and by the Center for Drug Delivery and Nanomedicine and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center (Omaha, NE). The speakers included established internationally regarded scientists from these institutions as well as graduate students and faculties at MSU. In addition to selected papers by workshop contributors, we have included several papers closely aligned to the theme of nanomedicine and nanopharmacology of the central nervous system in order to provide a biological anchor for this research. We understand that such works are new to many but hope that its organization and interdisciplinary approaches will appeal to this audience. All together, it is our hope that, by gathering basic and clinical scientists with the common interest of using nanotechnology in the delivery of therapeutic agents with a focus on nanopharmacology and complex supramolecular biological assembly, the papers included will provide a platform for thought, discussion, and future translational research.
Collapse
|
38
|
Keblesh JP, Reiner BC, Liu J, Xiong H. Pathogenesis of Human Immunodeficiency Virus Type-1 (HIV-1)-Associated Dementia: Role of Voltage-Gated Potassium Channels. RETROVIROLOGY : RESEARCH AND TREATMENT 2008; 2:1-10. [PMID: 20651955 PMCID: PMC2908044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
HIV-1-associated dementia (HAD) describes the cognitive impairments and behavioral disturbances which afflict many HIV-infected individuals. Although the incidence of HAD has decreased significantly in the era of HAART, it remains a significant complication of HIV-1 infection as patients with acquired immune deficient syndrome (AIDS) live longer, antiretroviral drugs remain unable to effectively cross the blood-brain barrier (BBB), and HIV-1 resistance grows due to viral strain mutation. Although the precise mechanism leading to HAD is incompletely understood, it is commonly accepted its progression involves a critical mass of infected and activated mononuclear phagocytes (MP; brain perivascular macrophages and microglia) releasing immune and viral products in brain. These cellular and viral products induce neuronal dysfunction and injury via various signaling pathways. Emerging evidence indicates that voltage-gated potassium (K(v)) channels, key regulators of cell excitability and animal behavior (learning and memory), are involved in the pathogenesis of HAD/HAND. Here we survey the literature and find HAD related alterations in cellular and viral products can alter MP and neuronal K(v) channel activity, leading to MP and neuronal dysfunction and cognitive deficits. Thus, MP and neuronal K(v) channels may be a new target in the effort to develop therapies for HAD and perhaps other inflammatory neurodegenerative disorders.
Collapse
|