1
|
Hu Z, Wu T, Zhou Z, Zhang Y, Chen Q, Yao H, Ji M, Shen G, Dong C, Shi C, Huang Z, Jiang N, Han N, Tian X. Asiaticoside Attenuates Blood-Spinal Cord Barrier Disruption by Inhibiting Endoplasmic Reticulum Stress in Pericytes After Spinal Cord Injury. Mol Neurobiol 2024; 61:678-692. [PMID: 37653222 DOI: 10.1007/s12035-023-03605-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
The blood-spinal cord barrier (BSCB) plays a vital role in the recovery of spinal cord function after spinal cord injury (SCI). Pericytes, pluripotent members of the neurovascular unit (NVU), receive signals from neighboring cells and are critical for maintaining CNS function. Therapeutic targets for the BSCB include endothelial cells (ECs) and glial cells, but few drugs target pericytes. This study was designed to explore whether asiaticoside has a positively effect on pericytes and the integrity of the BSCB. In this study, we found that asiaticoside could inhibit the loss of junction proteins just 1 day after SCI in vivo, but our in vitro study showed no significant differences in the expression of endothelial junction proteins between the control and asiaticoside treatment groups. We also found that asiaticoside could inhibit endoplasmic reticulum (ER) stress and pericyte apoptosis, which might be associated with the inhibition of junction protein reduction in ECs. Thus, we investigated the interactions between pericytes and ECs. Our results showed that asiaticoside could decrease the release of matrix metalloproteinase (MMP)-9 in pericytes and therefore upregulate the expression of junction proteins in ECs. Furthermore, the protective effect of asiaticoside on pericytes is related to the inhibition of ER stress via the MAPK signaling pathway. Taken together, our results demonstrate that asiaticoside treatment inhibits BSCB disruption and enhances functional recovery after SCI.
Collapse
Affiliation(s)
- Zhenxin Hu
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Tingting Wu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ziheng Zhou
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yu Zhang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315302, China
| | - Qiyue Chen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hanbing Yao
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Mengchu Ji
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ge Shen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chenling Dong
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chengge Shi
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhixian Huang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Nizhou Jiang
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Nan Han
- Department of Ultrasonography, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Xiliang Tian
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
2
|
Chen JTC, Hu X, Otto IUC, Schürger C, von Bieberstein BR, Doppler K, Krug SM, Hankir MK, Blasig R, Sommer C, Brack A, Blasig IE, Rittner HL. Myelin barrier breakdown, mechanical hypersensitivity, and painfulness in polyneuropathy with claudin-12 deficiency. Neurobiol Dis 2023; 185:106246. [PMID: 37527762 DOI: 10.1016/j.nbd.2023.106246] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/25/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND The blood-nerve and myelin barrier shield peripheral neurons and their axons. These barriers are sealed by tight junction proteins, which control the passage of potentially noxious molecules including proinflammatory cytokines via paracellular pathways. Peripheral nerve barrier breakdown occurs in various neuropathies, such as chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) and traumatic neuropathy. Here, we studied the functional role of the tight junction protein claudin-12 in regulating peripheral nerve barrier integrity and CIDP pathogenesis. METHODS Sections from sural nerve biopsies from 23 patients with CIDP and non-inflammatory idiopathic polyneuropathy (PNP) were analyzed for claudin-12 and -19 immunoreactivity. Cldn12-KO mice were generated and subjected to the chronic constriction injury (CCI) model of neuropathy. These mice were then characterized using a battery of barrier and behavioral tests, histology, immunohistochemistry, and mRNA/protein expression. In phenotype rescue experiments, the proinflammatory cytokine TNFα was neutralized with the anti-TNFα antibody etanercept; the peripheral nerve barrier was stabilized with the sonic hedgehog agonist smoothened (SAG). RESULTS Compared to those without pain, patients with painful neuropathy exhibited reduced claudin-12 expression independently of fiber loss. Accordingly, global Cldn12-KO in male mice, but not fertile female mice, selectively caused mechanical allodynia associated with a leaky myelin barrier, increased TNFα, decreased sonic hedgehog (SHH), and loss of small axons accompanied by reduced peripheral myelin protein 22 (Pmp22). Other barriers and neurological functions remained intact. The Cldn12-KO phenotype could be rescued either by neutralizing TNFα with etanercept or stabilizing the barrier with SAG, which both also upregulated the Schwann cell barrier proteins Cldn19 and Pmp22. CONCLUSION These results point to a critical role for claudin-12 in maintaining the myelin barrier presumably via Pmp22 and highlight restoration of the hedgehog pathway as a potential treatment strategy for painful inflammatory neuropathy.
Collapse
Affiliation(s)
- Jeremy Tsung-Chieh Chen
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Xiawei Hu
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Isabel U C Otto
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Christina Schürger
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Bruno Rogalla von Bieberstein
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Kathrin Doppler
- University Hospital Würzburg, Department of Neurology, 97080 Würzburg, Germany
| | - Susanne M Krug
- Charité-Universitätsmedizin Berlin, Clinical Physiology/Nutritional Medicine, 13125 Berlin, Germany
| | - Mohammed K Hankir
- University Hospital Würzburg, Department of General, Transplantation, Visceral, Vascular and Pediatric Surgery, 97080 Würzburg, Germany
| | - Rosel Blasig
- Leibnitz Institute of Molecular Pharmacology, Departments of Molecular Physiology and Cell Biology, 13125 Berlin, Germany
| | - Claudia Sommer
- University Hospital Würzburg, Department of Neurology, 97080 Würzburg, Germany
| | - Alexander Brack
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Ingolf E Blasig
- Leibnitz Institute of Molecular Pharmacology, Departments of Molecular Physiology and Cell Biology, 13125 Berlin, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany.
| |
Collapse
|
3
|
Reinhold AK, Hartmannsberger B, Burek M, Rittner HL. Stabilizing the neural barrier - A novel approach in pain therapy. Pharmacol Ther 2023; 249:108484. [PMID: 37390969 DOI: 10.1016/j.pharmthera.2023.108484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/08/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Chronic and neuropathic pain are a widespread burden. Incomplete understanding of underlying pathomechanisms is one crucial factor for insufficient treatment. Recently, impairment of the blood nerve barrier (BNB) has emerged as one key aspect of pain initiation and maintenance. In this narrative review, we discuss several mechanisms and putative targets for novel treatment strategies. Cells such as pericytes, local mediators like netrin-1 and specialized proresolving mediators (SPMs), will be covered as well as circulating factors including the hormones cortisol and oestrogen and microRNAs. They are crucial in either the BNB or similar barriers and associated with pain. While clinical studies are still scarce, these findings might provide valuable insight into mechanisms and nurture development of therapeutic approaches.
Collapse
Affiliation(s)
- Ann-Kristin Reinhold
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Beate Hartmannsberger
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Malgorzata Burek
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany.
| |
Collapse
|
4
|
Łach A, Wnuk A, Wójtowicz AK. Experimental Models to Study the Functions of the Blood-Brain Barrier. Bioengineering (Basel) 2023; 10:bioengineering10050519. [PMID: 37237588 DOI: 10.3390/bioengineering10050519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/07/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
The purpose of this paper was to discuss the achievements of in vitro modeling in terms of the blood-brain barrier [BBB] and to create a clear overview of this research area, which is useful in research planning. The text was divided into three main parts. The first part describes the BBB as a functional structure, its constitution, cellular and noncellular components, mechanisms of functioning and importance for the central nervous system, in terms of both protection and nourishment. The second part is an overview of parameters important in terms of establishing and maintaining a barrier phenotype that allows for formulating criteria of evaluation of the BBB in vitro models. The third and last part discusses certain techniques for developing the BBB in vitro models. It describes subsequent research approaches and models, as they underwent change alongside technological advancement. On the one hand, we discuss possibilities and limitations of different research approaches: primary cultures vs. cell lines and monocultures vs. multicultures. On the other hand, we review advantages and disadvantages of specific models, such as models-on-a-chip, 3D models or microfluidic models. We not only attempt to state the usefulness of specific models in different kinds of research on the BBB but also emphasize the significance of this area of research for advancement of neuroscience and the pharmaceutical industry.
Collapse
Affiliation(s)
- Andrzej Łach
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, 30-059 Kraków, Poland
| | - Agnieszka Wnuk
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Anna Katarzyna Wójtowicz
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, 30-059 Kraków, Poland
| |
Collapse
|
5
|
Sun Y, Zabihi M, Li Q, Li X, Kim BJ, Ubogu EE, Raja SN, Wesselmann U, Zhao C. Drug Permeability: From the Blood-Brain Barrier to the Peripheral Nerve Barriers. ADVANCED THERAPEUTICS 2023; 6:2200150. [PMID: 37649593 PMCID: PMC10465108 DOI: 10.1002/adtp.202200150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Indexed: 01/20/2023]
Abstract
Drug delivery into the peripheral nerves and nerve roots has important implications for effective local anesthesia and treatment of peripheral neuropathies and chronic neuropathic pain. Similar to drugs that need to cross the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) to gain access to the central nervous system (CNS), drugs must cross the peripheral nerve barriers (PNB), formed by the perineurium and blood-nerve barrier (BNB) to modulate peripheral axons. Despite significant progress made to develop effective strategies to enhance BBB permeability in therapeutic drug design, efforts to enhance drug permeability and retention in peripheral nerves and nerve roots are relatively understudied. Guided by knowledge describing structural, molecular and functional similarities between restrictive neural barriers in the CNS and peripheral nervous system (PNS), we hypothesize that certain CNS drug delivery strategies are adaptable for peripheral nerve drug delivery. In this review, we describe the molecular, structural and functional similarities and differences between the BBB and PNB, summarize and compare existing CNS and peripheral nerve drug delivery strategies, and discuss the potential application of selected CNS delivery strategies to improve efficacious drug entry for peripheral nerve disorders.
Collapse
Affiliation(s)
- Yifei Sun
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Mahmood Zabihi
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Qi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Xiaosi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Brandon J. Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa AL 35487, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham AL 35294, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa AL 35487, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa AL 35487, USA
| | - Eroboghene E. Ubogu
- Division of Neuromuscular Disease, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Srinivasa N. Raja
- Division of Pain Medicine, Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Ursula Wesselmann
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, and Department of Neurology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Consortium for Neuroengineering and Brain-Computer Interfaces, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa AL 35487, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa AL 35487, USA
| |
Collapse
|
6
|
IWANAGA T, TAKAHASHI-IWANAGA H, NIO-KOBAYASHI J, EBARA S. Structure and barrier functions of the perineurium and its relationship with associated sensory corpuscles: A review. Biomed Res 2022; 43:145-159. [DOI: 10.2220/biomedres.43.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Toshihiko IWANAGA
- Department of Anatomy, Hokkaido University Graduate School of Medicine
| | | | | | | |
Collapse
|
7
|
In vitro blood brain barrier models: An overview. J Control Release 2022; 343:13-30. [PMID: 35026351 DOI: 10.1016/j.jconrel.2022.01.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/22/2022]
Abstract
Understanding the composition and function of the blood brain barrier (BBB) enables the development of novel, innovative techniques for administering central nervous system (CNS) medications and technologies for improving the existing models. Scientific and methodological interest in the pathology of the BBB resulted in the formation of numerous in vitro BBB models. Once successfully studied and modelled, it would be a valuable tool for elucidating the mechanism of action of the CNS disorders prior to their manifestation and the pathogenic factors. Understanding the rationale behind the selection of the models as well as their working may enable the development of state-of-the-art drugs for treating and managing neurological diseases. Hence, to have realistic simulation of the BBB and test its drug permeability the microfluidics-based BBB-on-Chip model has been developed. To summarise, we aim to evaluate the advanced, newly developed and frequently used in vitro BBB models, thereby providing a brief overview of the components essential for in vitro BBB formation, the methods of chip fabrication and cell culturing, its applications and the recent advances in this technological field. This will be critical for developing CNS treatments with improved BBB penetrability and pharmacokinetic properties.
Collapse
|
8
|
Bhargavan B, Woollard SM, McMillan JE, Kanmogne GD. CCR5 antagonist reduces HIV-induced amyloidogenesis, tau pathology, neurodegeneration, and blood-brain barrier alterations in HIV-infected hu-PBL-NSG mice. Mol Neurodegener 2021; 16:78. [PMID: 34809709 PMCID: PMC8607567 DOI: 10.1186/s13024-021-00500-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neurocognitive impairment is present in 50% of HIV-infected individuals and is often associated with Alzheimer's Disease (AD)-like brain pathologies, including increased amyloid-beta (Aβ) and Tau hyperphosphorylation. Here, we aimed to determine whether HIV-1 infection causes AD-like pathologies in an HIV/AIDS humanized mouse model, and whether the CCR5 antagonist maraviroc alters HIV-induced pathologies. METHODS NOD/scid-IL-2Rγcnull mice engrafted with human blood leukocytes were infected with HIV-1, left untreated or treated with maraviroc (120 mg/kg twice/day). Human cells in animal's blood were quantified weekly by flow cytometry. Animals were sacrificed at week-3 post-infection; blood and tissues viral loads were quantified using p24 antigen ELISA, RNAscope, and qPCR. Human (HLA-DR+) cells, Aβ-42, phospho-Tau, neuronal markers (MAP 2, NeuN, neurofilament-L), gamma-secretase activating protein (GSAP), and blood-brain barrier (BBB) tight junction (TJ) proteins expression and transcription were quantified in brain tissues by immunohistochemistry, immunofluorescence, immunoblotting, and qPCR. Plasma Aβ-42, Aβ-42 cellular uptake, release and transendothelial transport were quantified by ELISA. RESULTS HIV-1 significantly decreased human (h)CD4+ T-cells and hCD4/hCD8 ratios; decreased the expression of BBB TJ proteins claudin-5, ZO-1, ZO-2; and increased HLA-DR+ cells in brain tissues. Significantly, HIV-infected animals showed increased plasma and brain Aβ-42 and phospho-Tau (threonine181, threonine231, serine396, serine199), associated with transcriptional upregulation of GSAP, an enzyme that catalyzes Aβ formation, and loss of MAP 2, NeuN, and neurofilament-L. Maraviroc treatment significantly reduced blood and brain viral loads, prevented HIV-induced loss of neuronal markers and TJ proteins; decreased HLA-DR+ cells infiltration in brain tissues, significantly reduced HIV-induced increase in Aβ-42, GSAP, and phospho-Tau. Maraviroc also reduced Aβ retention and increased Aβ release in human macrophages; decreased the receptor for advanced glycation end products (RAGE) and increased low-density lipoprotein receptor-related protein-1 (LRP1) expression in human brain endothelial cells. Maraviroc induced Aβ transendothelial transport, which was blocked by LRP1 antagonist but not RAGE antagonist. CONCLUSIONS Maraviroc significantly reduced HIV-induced amyloidogenesis, GSAP, phospho-Tau, neurodegeneration, BBB alterations, and leukocytes infiltration into the CNS. Maraviroc increased cellular Aβ efflux and transendothelial Aβ transport via LRP1 pathways. Thus, therapeutically targeting CCR5 could reduce viremia, preserve the BBB and neurons, increased brain Aβ efflux, and reduce AD-like neuropathologies.
Collapse
Affiliation(s)
- Biju Bhargavan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| | - Shawna M. Woollard
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
- Huvepharma, 421 W Industrial Lake Drive, Lincoln, NE 68528 USA
| | - Jo Ellyn McMillan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| | - Georgette D. Kanmogne
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| |
Collapse
|
9
|
Takeshita Y, Fujikawa S, Serizawa K, Fujisawa M, Matsuo K, Nemoto J, Shimizu F, Sano Y, Tomizawa-Shinohara H, Miyake S, Ransohoff RM, Kanda T. New BBB Model Reveals That IL-6 Blockade Suppressed the BBB Disorder, Preventing Onset of NMOSD. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1076. [PMID: 34667128 PMCID: PMC8529420 DOI: 10.1212/nxi.0000000000001076] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/24/2021] [Indexed: 01/04/2023]
Abstract
Background and Objectives To evaluate the pathophysiology of neuromyelitis optica spectrum disorder (NMOSD) and the therapeutic mechanism and levels of interleukin-6 (IL-6) blockade (satralizumab), especially with respect to blood-brain barrier (BBB) disruption with the new in vitro and ex vivo human BBB models and in vivo model. Methods We constructed new static in vitro and flow-based ex vivo models for evaluating continued barrier function, leukocyte transmigration, and intracerebral transferability of neuromyelitis optica-immunoglobulin G (NMO-IgG) and satralizumab across the BBB using the newly established triple coculture system that are specialized to closely mimic endothelial cell contact of pericytes and endfeet of astrocytes. In the in vivo study, we assessed the effects of an anti–IL-6 receptor antibody for mice (MR16-1) on in vivo BBB disruption in mice with experimental autoimmune encephalomyelitis in which IL-6 concentration in the spinal cord dramatically increases. Results In vitro and ex vivo experiments demonstrated that NMO-IgG increased intracerebral transferability of satralizumab and NMO-IgG and that satralizumab suppressed the NMO-IgG–induced transmigration of T cells and barrier dysfunction. In the in vivo study, the blockade of IL-6 signaling suppressed the migration of T cells into the spinal cord and prevented the increased BBB permeability. Discussion These results suggest that (1) our triple-cultured in vitro and in ex vivo BBB models are ideal for evaluating barrier function, leukocyte transmigration, and intracerebral transferability; (2) NMO-IgG increased the intracerebral transferability of NMO-IgG via decreasing barrier function and induced secretion of IL-6 from astrocytes causing more dysfunction of the barrier and disrupting controlled cellular infiltration; and (3) satralizumab, which can pass through the BBB in the presence of NMO-IgG, suppresses the BBB dysfunction and the infiltration of inflammatory cells, leading to prevention of onset of NMOSD.
Collapse
Affiliation(s)
- Yukio Takeshita
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Susumu Fujikawa
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Kenichi Serizawa
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Miwako Fujisawa
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Kinya Matsuo
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Joe Nemoto
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Fumitaka Shimizu
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Yasuteru Sano
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Haruna Tomizawa-Shinohara
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Shota Miyake
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Richard M Ransohoff
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA
| | - Takashi Kanda
- From the Department of Neurology and Clinical Neuroscience (Y.T., S.F., M.F., K.M., J.N., F.S., Y.S., T.K.), Yamaguchi University Graduate School of Medicine; Kenichi Serizawa (K.S., H.T.-S., S.M.), Haruna Tomizawa-Shinohara and Shota Miyake, Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan; and Richard M Ransohoff (R.M.R.), Third Rock Ventures, Boston, MA.
| |
Collapse
|
10
|
Zhang Z, Lu Z, Liu C, Man J, Li X, Cui K, Lu H, Wang J. Protective effects of Dimethyl malonate on neuroinflammation and blood-brain barrier after ischemic stroke. Neuroreport 2021; 32:1161-1169. [PMID: 34334775 DOI: 10.1097/wnr.0000000000001704] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES After ischemic stroke, microglia will be activated and play a key role in neuroinflammation and the destruction of the blood-brain barrier (BBB), and activated microglia could polarize into pro-inflammation M1 phenotype and anti-inflammation M2 phenotype. Dimethyl malonate (DMM) could reduce reactive oxygen species and we speculate DMM could regulate microglia to protect ischemic brain. METHODS We used transient middle cerebral artery occlusion (tMCAO) mouse model to simulate ischemic stroke and adult male C57BL/6 mice were used in our study. 2,3,5-triphenyltetrazolium chloride staining was used to measure infarct volume. Evans Blue and Brain water content were used to evaluate the destruction of BBB. We used a five-point scale to assess the neurologic function of mice. Western blot and Immunofluorescence were used to measure microglia, pericytes and the expression of related proteins. RESULTS DMM reduced cerebral infarct volume, Evans blue leakage, brain water content and improved neurologic deficits after tMCAO. The number of activated microglia and M1 microglia were decreased and the number of M2 microglia and pericytes were increased after DMM treatment. The expression of tumor necrosis factor-α was reduced while protein levels of IL-10 and ZO-1 were increased through DMM treatment. CONCLUSIONS DMM could regulate activation and polarization of microglia to inhibit neuroinflammation and protect BBB.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University
| | - Zhengfang Lu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University
| | - Chang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University
| | - Jiang Man
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University
| | - Xiang Li
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University
| | | | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianping Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University
| |
Collapse
|
11
|
Ran X, Zhang Q, Li S, Yu Z, Wan L, Wu B, Wu R, Li S. Tissue Kallikrein Exacerbating Sepsis-Induced Endothelial Hyperpermeability is Highly Predictive of Severity and Mortality in Sepsis. J Inflamm Res 2021; 14:3321-3333. [PMID: 34290517 PMCID: PMC8289368 DOI: 10.2147/jir.s317874] [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: 05/04/2021] [Accepted: 07/03/2021] [Indexed: 11/23/2022] Open
Abstract
Aim Sepsis, an acute, life-threatening dysregulated response to infection, affects practically all aspects of endothelial function. Tissue kallikrein (TK) is a key enzyme in the kallikrein–kinin system (KKS) which has been implicated in endothelial permeability. Thus, we aimed to establish a potentially novel association among TK, endothelial permeability, and sepsis demonstrated by clinical investigation and in vitro studies. Methods We performed a clinical investigation with the participation of a total of 76 controls, 42 systemic inflammatory response syndrome (SIRS) patients, and 150 patients with sepsis, who were followed-up for 28 days. Circulating TK levels were measured with an enzyme-linked immunosorbent assay. Then, the effect of TK on sepsis-induced endothelial hyperpermeability was evaluated by in vitro study. Results Data showed a gradual increase in TK level among controls and the patients with SIRS, sepsis, and septic shock (0.288±0.097 mg/l vs 0.335±0.149 vs 0.495±0.170 vs 0.531±0.188 mg/l, respectively, P <0.001). Further analysis revealed that plasma TK level was positively associated with the severity and mortality of sepsis and negatively associated with event-free survival during 28 days of follow-up (relative risk, 3.333; 95% CI, 2.255–4.925; p < 0.001). With a septic model of TK and kallistatin in vitro, we found that TK exacerbated sepsis-induced endothelial hyperpermeability by downregulating zonula occluden-1 (ZO-1) and vascular endothelial (VE)-cadherin, and these could be reversed by kallistatin, an inhibitor of TK. Conclusion TK can be used in the diagnosis of sepsis and assessment of severity and prognosis of disease. Inhibition of TK may be a novel therapeutic target for sepsis through increasing ZO-1 and VE-cadherin, as well as downregulating endothelial permeability.
Collapse
Affiliation(s)
- Xiao Ran
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Qin Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Shaoping Li
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Zhen Yu
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Li Wan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Bin Wu
- Laboratory of Platelet and Endothelium Biology, Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Rongxue Wu
- Department of Biological Sciences Division/Cardiology, University of Chicago, Chicago, IL, 60637, USA
| | - Shusheng Li
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| |
Collapse
|
12
|
Scalise AA, Kakogiannos N, Zanardi F, Iannelli F, Giannotta M. The blood-brain and gut-vascular barriers: from the perspective of claudins. Tissue Barriers 2021; 9:1926190. [PMID: 34152937 PMCID: PMC8489939 DOI: 10.1080/21688370.2021.1926190] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In some organs, such as the brain, endothelial cells form a robust and highly selective blood-to-tissue barrier. However, in other organs, such as the intestine, endothelial cells provide less stringent permeability, to allow rapid exchange of solutes and nutrients where needed. To maintain the structural and functional integrity of the highly dynamic blood–brain and gut–vascular barriers, endothelial cells form highly specialized cell-cell junctions, known as adherens junctions and tight junctions. Claudins are a family of four-membrane-spanning proteins at tight junctions and they have both barrier-forming and pore-forming properties. Tissue-specific expression of claudins has been linked to different diseases that are characterized by barrier impairment. In this review, we summarize the more recent progress in the field of the claudins, with particular attention to their expression and function in the blood–brain barrier and the recently described gut–vascular barrier, under physiological and pathological conditions. Abbreviations: 22q11DS 22q11 deletion syndrome; ACKR1 atypical chemokine receptor 1; AD Alzheimer disease; AQP aquaporin; ATP adenosine triphosphate; Aβ amyloid β; BAC bacterial artificial chromosome; BBB blood-brain barrier; C/EBP-α CCAAT/enhancer-binding protein α; cAMP cyclic adenosine monophosphate (or 3ʹ,5ʹ-cyclic adenosine monophosphate); CD cluster of differentiation; CNS central nervous system; DSRED discosoma red; EAE experimental autoimmune encephalomyelitis; ECV304 immortalized endothelial cell line established from the vein of an apparently normal human umbilical cord; EGFP enhanced green fluorescent protein; ESAM endothelial cell-selective adhesion molecule; GLUT-1 glucose transporter 1; GVB gut-vascular barrier; H2B histone H2B; HAPP human amyloid precursor protein; HEK human embryonic kidney; JACOP junction-associated coiled coil protein; JAM junctional adhesion molecules; LYVE1 lymphatic vessel endothelial hyaluronan receptor 1; MADCAM1 mucosal vascular addressin cell adhesion molecule 1; MAPK mitogen-activated protein kinase; MCAO middle cerebral artery occlusion; MMP metalloprotease; MS multiple sclerosis; MUPP multi-PDZ domain protein; PATJ PALS-1-associated tight junction protein; PDGFR-α platelet-derived growth factor receptor α polypeptide; PDGFR-β platelet-derived growth factor receptor β polypeptide; RHO rho-associated protein kinase; ROCK rho-associated, coiled-coil-containing protein kinase; RT-qPCR real time quantitative polymerase chain reactions; PDGFR-β soluble platelet-derived growth factor receptor, β polypeptide; T24 human urinary bladder carcinoma cells; TG2576 transgenic mice expressing the human amyloid precursor protein; TNF-α tumor necrosis factor α; WTwild-type; ZO zonula occludens.
Collapse
|
13
|
Wang Y, Zhang F, Xiong N, Xu H, Chai S, Wang H, Wang J, Zhao H, Jiang X, Fu P, Xiang W. Remodelling and Treatment of the Blood-Brain Barrier in Glioma. Cancer Manag Res 2021; 13:4217-4232. [PMID: 34079374 PMCID: PMC8166259 DOI: 10.2147/cmar.s288720] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/30/2021] [Indexed: 11/23/2022] Open
Abstract
The blood-brain barrier (BBB) is an essential structure of the central nervous system (CNS), and its existence makes the local internal environment of the CNS a relatively independent structure distinct from other internal environments of the human body to ensure normal physiological and high stability of activities of the CNS. Changes in BBB structure and function are fundamental to the pathophysiology of many diseases. The occurrence and development of glioma are often accompanied by a series of changes in the structure and function of the internal environment, the most significant of which is remodelling of the BBB. The remodelling of the BBB usually leads to changes in the permeability of local microvessels, which provide certain favourable conditions for the occurrence and development of glioma. Meanwhile, the newly generated abnormal blood vessels and the remaining intact regions of the BBB also hinder the effects of drug treatments. Changes in permeability and structural function often lead to the creation of abnormally functioning vascular regions, which pose further treatment challenges. At present, therapeutic methods for glioma have not achieved satisfactory effects in clinical practice, and emerging therapeutic methods have not yet been widely used in clinical practice. In this review, we summarize the knowledge of remodelling of the BBB in the glioma environment, the type of changes that occur, and current BBB treatment methods and prospects for the treatment of glioma.
Collapse
Affiliation(s)
- Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Fangcheng Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Nanxiang Xiong
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Hao Xu
- Department of Neurosurgery, General Hospital of the Yangtze River Shipping, Wuhan, 430022, People's Republic of China
| | - Songshan Chai
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jiajing Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Hongyang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
14
|
Surface Functionalization of PLGA Nanoparticles to Increase Transport across the BBB for Alzheimer’s Disease. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11094305] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disorder that accounts for about 60% of all diagnosed cases of dementia worldwide. Although there are currently several drugs marketed for its treatment, none are capable of slowing down or stopping the progression of AD. The role of the blood-brain barrier (BBB) plays a key role in the design of a successful treatment for this neurodegenerative disease. Nanosized particles have been proposed as suitable drug delivery systems to overcome BBB with the purpose of increasing bioavailability of drugs in the brain. Biodegradable poly (lactic-co-glycolic acid) nanoparticles (PLGA-NPs) have been particularly regarded as promising drug delivery systems as they can be surface-tailored with functionalized molecules for site-specific targeting. In this review, a thorough discussion about the most recent functionalization strategies based on PLGA-NPs for AD and their mechanisms of action is provided, together with a description of AD pathogenesis and the role of the BBB in brain targeting.
Collapse
|
15
|
Frías-Anaya E, Gromnicova R, Kraev I, Rogachevsky V, Male DK, Crea F, Hawkes CA, Romero IA. Age-related ultrastructural neurovascular changes in the female mouse cortex and hippocampus. Neurobiol Aging 2021; 101:273-284. [PMID: 33579556 DOI: 10.1016/j.neurobiolaging.2020.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/29/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Blood-brain barrier (BBB) breakdown occurs in aging and neurodegenerative diseases. Although age-associated alterations have previously been described, most studies focused in male brains; hence, little is known about BBB breakdown in females. This study measured ultrastructural features in the aging female BBB using transmission electron microscopy and 3-dimensional reconstruction of cortical and hippocampal capillaries from 6- and 24-month-old female C57BL/6J mice. Aged cortical capillaries showed more changes than hippocampal capillaries. Specifically, the aged cortex showed thicker basement membrane, higher number and volume of endothelial pseudopods, decreased endothelial mitochondrial number, larger pericyte mitochondria, higher pericyte-endothelial cell contact, and increased tight junction tortuosity compared with young animals. Only increased basement membrane thickness and pericyte mitochondrial volume were observed in the aged hippocampus. Regional comparison revealed significant differences in endothelial pseudopods and tight junctions between the cortex and hippocampus of 24-month-old mice. Therefore, the aging female BBB shows region-specific ultrastructural alterations that may lead to oxidative stress and abnormal capillary blood flow and barrier stability, potentially contributing to cerebrovascular diseases, particularly in postmenopausal women.
Collapse
Affiliation(s)
- Eduardo Frías-Anaya
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Radka Gromnicova
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Igor Kraev
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Vadim Rogachevsky
- Institute of Cell Biophysics RAS, Pushchino Federal Research Centre for Biological Research, Pushchino, Russia
| | - David K Male
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Francesco Crea
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Cheryl A Hawkes
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK; Department of Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | - Ignacio A Romero
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
16
|
Zhang Q, Zheng M, Betancourt CE, Liu L, Sitikov A, Sladojevic N, Zhao Q, Zhang JH, Liao JK, Wu R. Increase in Blood-Brain Barrier (BBB) Permeability Is Regulated by MMP3 via the ERK Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6655122. [PMID: 33859779 PMCID: PMC8026308 DOI: 10.1155/2021/6655122] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/24/2020] [Accepted: 03/09/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The blood-brain barrier (BBB) regulates the exchange of molecules between the brain and peripheral blood and is composed primarily of microvascular endothelial cells (BMVECs), which form the lining of cerebral blood vessels and are linked via tight junctions (TJs). The BBB is regulated by components of the extracellular matrix (ECM), and matrix metalloproteinase 3 (MMP3) remodels the ECM's basal lamina, which forms part of the BBB. Oxidative stress is implicated in activation of MMPs and impaired BBB. Thus, we investigated whether MMP3 modulates BBB permeability. METHODS Experiments included in vivo assessments of isoflurane anesthesia and dye extravasation from brain in wild-type (WT) and MMP3-deficient (MMP3-KO) mice, as well as in vitro assessments of the integrity of monolayers of WT and MMP3-KO BMVECs and the expression of junction proteins. RESULTS Compared to WT mice, measurements of isoflurane usage and anesthesia induction time were higher in MMP3-KO mice and lower in WT that had been treated with MMP3 (WT+MMP3), while anesthesia emergence times were shorter in MMP3-KO mice and longer in WT+MMP3 mice than in WT. Extravasation of systemically administered dyes was also lower in MMP3-KO mouse brains and higher in WT+MMP3 mouse brains, than in the brains of WT mice. The results from both TEER and Transwell assays indicated that MMP3 deficiency (or inhibition) increased, while MMP3 upregulation reduced barrier integrity in either BMVEC or the coculture. MMP3 deficiency also increased the abundance of TJs and VE-cadherin proteins in BMVECs, and the protein abundance declined when MMP3 activity was upregulated in BMVECs, but not when the cells were treated with an inhibitor of extracellular signal related-kinase (ERK). CONCLUSION MMP3 increases BBB permeability following the administration of isoflurane by upregulating the ERK signaling pathway, which subsequently reduces TJ and VE-cadherin proteins in BMVECs.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Mei Zheng
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
| | | | - Lifeng Liu
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
| | - Albert Sitikov
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
| | - Nikola Sladojevic
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
| | - Qiong Zhao
- Division of Cardiology, Department of Medicine, Inova Heart and Vascular Institute, USA
| | - John H. Zhang
- Center for Neuroscience Research, Loma Linda University, School of Medicine, USA
| | - James K. Liao
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
| | - Rongxue Wu
- Department of Biological Sciences Division-Cardiology, University of Chicago, USA
| |
Collapse
|
17
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
18
|
Girolamo F, de Trizio I, Errede M, Longo G, d’Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021. [DOI: 10.1186/s12987-021-00242-7 union select null--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractCentral nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches.
Collapse
|
19
|
Blood-Nerve Barrier (BNB) Pathology in Diabetic Peripheral Neuropathy and In Vitro Human BNB Model. Int J Mol Sci 2020; 22:ijms22010062. [PMID: 33374622 PMCID: PMC7793499 DOI: 10.3390/ijms22010062] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022] Open
Abstract
In diabetic peripheral neuropathy (DPN), metabolic disorder by hyperglycemia progresses in peripheral nerves. In addition to the direct damage to peripheral neural axons, the homeostatic mechanism of peripheral nerves is disrupted by dysfunction of the blood–nerve barrier (BNB) and Schwann cells. The disruption of the BNB, which is a crucial factor in DPN development and exacerbation, causes axonal degeneration via various pathways. Although many reports revealed that hyperglycemia and other important factors, such as dyslipidemia-induced dysfunction of Schwann cells, contributed to DPN, the molecular mechanisms underlying BNB disruption have not been sufficiently elucidated, mainly because of the lack of in vitro studies owing to difficulties in establishing human cell lines from vascular endothelial cells and pericytes that form the BNB. We have developed, for the first time, temperature-sensitive immortalized cell lines of vascular endothelial cells and pericytes originating from the BNB of human sciatic nerves, and we have elucidated the disruption to the BNB mainly in response to advanced glycation end products in DPN. Recently, we succeeded in developing an in vitro BNB model to reflect the anatomical characteristics of the BNB using cell sheet engineering, and we established immortalized cell lines originating from the human BNB. In this article, we review the pathologic evidence of the pathology of DPN in terms of BNB disruption, and we introduce the current in vitro BNB models.
Collapse
|
20
|
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: 685] [Impact Index Per Article: 171.3] [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
|
21
|
Medina-Flores F, Hurtado-Alvarado G, Contis-Montes de Oca A, López-Cervantes SP, Konigsberg M, Deli MA, Gómez-González B. Sleep loss disrupts pericyte-brain endothelial cell interactions impairing blood-brain barrier function. Brain Behav Immun 2020; 89:118-132. [PMID: 32485292 DOI: 10.1016/j.bbi.2020.05.077] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/12/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022] Open
Abstract
Sleep loss in the rat increases blood-brain barrier permeability to circulating molecules by disrupting interendothelial tight junctions. Despite the description of the ultrastructure of cerebral microvessels and the evidence of an apparent pericyte detachment from capillary wall in sleep restricted rats the effect of sleep loss on pericytes is unknown. Here we characterized the interactions between pericytes and brain endothelial cells after sleep loss using male Wistar rats. Animals were sleep-restricted 20 h daily with 4 h sleep recovery for 10 days. At the end of the sleep restriction, brain microvessels (MVs) were isolated from cerebral cortex and hippocampus and processed for Western blot and immunocytochemistry to evaluate markers of pericyte-endothelial cell interaction (connexin 43, PDGFR-β), tight junction proteins, and proinflammatory mediator proteins (MMP9, A2A adenosine receptor, CD73, NFκB). Sleep restriction reduced PDGFR-β and connexin 43 expression in MVs; in addition, scanning electron microscopy micrographs showed that pericytes were detached from capillary walls, but did not undergo apoptosis (as depicted by a reduced active caspase-3 expression). Sleep restriction also decreased tight junction protein expression in MVs and increased BBB permeability to low- and high-molecular weight tracers in in vivo permeability assays. Those alterations seemed to depend on a low-grade inflammatory status as reflected by the increased expression of phosphorylated NFκB and A2A adenosine receptor in brain endothelial cells from the sleep-restricted rats. Our data show that pericyte-brain endothelial cell interaction is altered by sleep restriction; this evidence is essential to understand the role of sleep in regulating blood-brain barrier function.
Collapse
Affiliation(s)
- Fernanda Medina-Flores
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Gabriela Hurtado-Alvarado
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Arturo Contis-Montes de Oca
- Universidad Nacional Autónoma de México (UNAM), Facultad de Estudios Superiores (FES) Iztacala, Optometría, Mexico
| | - Stefanie Paola López-Cervantes
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico; Laboratorio de Bioenergética y Envejecimiento Celular, Dept. Health Sciences, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico D.F., Mexico
| | - Mina Konigsberg
- Laboratorio de Bioenergética y Envejecimiento Celular, Dept. Health Sciences, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico D.F., Mexico.
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary; Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary.
| | - Beatriz Gómez-González
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico.
| |
Collapse
|
22
|
Stubbs EB. Targeting the blood-nerve barrier for the management of immune-mediated peripheral neuropathies. Exp Neurol 2020; 331:113385. [PMID: 32562668 DOI: 10.1016/j.expneurol.2020.113385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
Healthy peripheral nerves encounter, with increased frequency, numerous chemical, biological, and biomechanical forces. Over time and with increasing age, these forces collectively contribute to the pathophysiology of a spectrum of traumatic, metabolic, and/or immune-mediated peripheral nerve disorders. The blood-nerve barrier (BNB) serves as a critical first-line defense against chemical and biologic insults while biomechanical forces are continuously buffered by a dense array of longitudinally orientated epineural collagen fibers exhibiting high-tensile strength. As emphasized throughout this Experimental Neurology Special Issue, the BNB is best characterized as a functionally dynamic multicellular vascular unit comprised of not only highly specialized endoneurial endothelial cells, but also associated perineurial cells, pericytes, Schwann cells, basement membrane, and invested axons. The composition of the BNB, while anatomically distinct, is not functionally dissimilar to that of the well characterized neurovascular unit of the central nervous system. While the BNB lacks a glial limitans and an astrocytic endfoot layer, the primary function of both vascular units is to establish, maintain, and protect an optimal endoneurial (PNS) or interstitial (CNS) fluid microenvironment that is vital for proper neuronal function. Altered endoneurial homeostasis as a secondary consequence of BNB dysregulation is considered an early pathological event in the course of a variety of traumatic, immune-mediated, or metabolically acquired peripheral neuropathies. In this review, emerging experimental advancements targeting the endoneurial microvasculature for the therapeutic management of immune-mediated inflammatory peripheral neuropathies, including the AIDP variant of Guillain-Barré syndrome, are discussed.
Collapse
Affiliation(s)
- Evan B Stubbs
- Research Service (151), Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL 60141, USA; Department of Ophthalmology, Loyola University Health Science Division, Maywood, IL 60153, USA.
| |
Collapse
|
23
|
Deligne C, Hachani J, Duban-Deweer S, Meignan S, Leblond P, Carcaboso AM, Sano Y, Shimizu F, Kanda T, Gosselet F, Dehouck MP, Mysiorek C. Development of a human in vitro blood-brain tumor barrier model of diffuse intrinsic pontine glioma to better understand the chemoresistance. Fluids Barriers CNS 2020; 17:37. [PMID: 32487241 PMCID: PMC7268424 DOI: 10.1186/s12987-020-00198-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/23/2020] [Indexed: 02/08/2023] Open
Abstract
Background Pediatric diffuse intrinsic pontine glioma (DIPG) represents one of the most devastating and lethal brain tumors in children with a median survival of 12 months. The high mortality rate can be explained by the ineligibility of patients to surgical resection due to the diffuse growth pattern and midline localization of the tumor. While the therapeutic strategies are unfortunately palliative, the blood–brain barrier (BBB) is suspected to be responsible for the treatment inefficiency. Located at the brain capillary endothelial cells (ECs), the BBB has specific properties to tightly control and restrict the access of molecules to the brain parenchyma including chemotherapeutic compounds. However, these BBB specific properties can be modified in a pathological environment, thus modulating brain exposure to therapeutic drugs. Hence, this study aimed at developing a syngeneic human blood–brain tumor barrier model to understand how the presence of DIPG impacts the structure and function of brain capillary ECs. Methods A human syngeneic in vitro BBB model consisting of a triple culture of human (ECs) (differentiated from CD34+-stem cells), pericytes and astrocytes was developed. Once validated in terms of BBB phenotype, this model was adapted to develop a blood–brain tumor barrier (BBTB) model specific to pediatric DIPG by replacing the astrocytes by DIPG-007, -013 and -014 cells. The physical and metabolic properties of the BBTB ECs were analyzed and compared to the BBB ECs. The permeability of both models to chemotherapeutic compounds was evaluated. Results In line with clinical observation, the integrity of the BBTB ECs remained intact until 7 days of incubation. Both transcriptional expression and activity of efflux transporters were not strongly modified by the presence of DIPG. The permeability of ECs to the chemotherapeutic drugs temozolomide and panobinostat was not affected by the DIPG environment. Conclusions This original human BBTB model allows a better understanding of the influence of DIPG on the BBTB ECs phenotype. Our data reveal that the chemoresistance described for DIPG does not come from the development of a “super BBB”. These results, validated by the absence of modification of drug transport through the BBTB ECs, point out the importance of understanding the implication of the different protagonists in the pathology to have a chance to significantly improve treatment efficiency.
Collapse
Affiliation(s)
- Clémence Deligne
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, UR 2465, 62300, Lens, France
| | - Johan Hachani
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Plateau Spectrométrie de Masse de l'ARTois (SMART), Univ. Artois, UR 2465, 62300, Lens, France
| | - Sophie Duban-Deweer
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Plateau Spectrométrie de Masse de l'ARTois (SMART), Univ. Artois, UR 2465, 62300, Lens, France
| | - Samuel Meignan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U908, 59000, Lille, France.,Institut pour la Recherche sur le Cancer de Lille (IRCL), 59000, Lille, France.,Unité Tumorigenèse et Résistance aux Traitements, Centre Oscar Lambret, 3 rue Frédéric Combemale, 59000, Lille, France
| | - Pierre Leblond
- Département de Cancérologie pédiatrique, Institut d'Hématologie et d'Oncologie Pédiatrique, 69000, Lyon, France
| | - Angel M Carcaboso
- Institut de Recerca Sant Joan de Deu, Esplugues de Llobregat, 08950, Barcelona, Spain
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, UR 2465, 62300, Lens, France
| | - Marie-Pierre Dehouck
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, UR 2465, 62300, Lens, France
| | - Caroline Mysiorek
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, UR 2465, 62300, Lens, France.
| |
Collapse
|
24
|
Revisiting the blood-brain barrier: A hard nut to crack in the transportation of drug molecules. Brain Res Bull 2020; 160:121-140. [PMID: 32315731 DOI: 10.1016/j.brainresbull.2020.03.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022]
Abstract
Barriers are the hallmark of a healthy physiology, blood-brain barrier (BBB) being a tough nut to crack for most of the antigens and chemical substances. The presence of tight junctions plays a remarkable role in defending the brain from antigenic and pathogenic attacks. BBB constitutes a diverse assemblage of multiple physical and chemical barriers that judiciously restrict the flux of blood solutes into and out of the brain. Restrictions through the paracellular pathway and the tight junctions between intercellular clefts, together create well regulated metabolic and transport barricades, critical to brain pathophysiology. The brain being impermeable to many essential metabolites and nutrients regulates transportation via specialized transport systems across the endothelial abluminal and luminal membranes. The epithelial cells enveloping capillaries of the choroid plexus regulates the transport of complement, growth factors, hormones, microelements, peptides and trace elements into ventricles. Nerve terminals, microglia, and pericytes associated with the endothelium support barrier induction and function, ensuring an optimally stable ionic microenvironment that facilitates neurotransmission, orchestrated by multiple ion channels (Na+, K+ Mg2+, Ca2+) and transporters. Brain pathology which can develop due to genetic mutations or secondary to other cerebrovascular, neurodegenerative diseases can cause aberration in the microvasculature of CNS which is the uniqueness of BBB. This can also alter BBB permeation and result in BBB breakdown and other neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The concluding section outlines contemporary trends in drug discovery, focusing on molecular determinants of BBB permeation and novel drug-delivery systems, such as dendrimers, liposomes, nanoparticles, nanogels, etc.
Collapse
|
25
|
Cyclophilin a signaling induces pericyte-associated blood-brain barrier disruption after subarachnoid hemorrhage. J Neuroinflammation 2020; 17:16. [PMID: 31926558 PMCID: PMC6954572 DOI: 10.1186/s12974-020-1699-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Objective The potential roles and mechanisms of pericytes in maintaining blood–brain barrier (BBB) integrity, which would be helpful for the development of therapeutic strategies for subarachnoid hemorrhage (SAH), remain unclear. We sought to provide evidence on the potential role of pericytes in BBB disruption and possible involvement and mechanism of CypA signaling in both cultured pericytes and SAH models. Methods Three hundred fifty-three adult male C57B6J mice weighing 22 to 30 g, 29 CypA−/− mice, 30 CypA+/+ (flox/flox) mice, and 30 male neonatal C57B6J mice were used to investigate the time course of CypA expression in pericytes after SAH, the intrinsic function and mechanism of CypA in pericytes, and whether the known receptor CD147 mediates these effects. Results Our data demonstrated both intracellular CypA and CypA secretion increased after SAH and could activate CD147 receptor and downstream NF-κB pathway to induce MMP9 expression and proteolytic functions for degradation of endothelium tight junction proteins and basal membranes. CypA served as autocrine or paracrine ligand for its receptor, CD147. Although CypA could be endocytosed by pericytes, specific endocytosis inhibitor chlorpromazine did not have any effect on MMP9 activation. However, specific knockdown of CD147 could reverse the harmful effects of CypA expression in pericytes on the BBB integrity after SAH. Conclusions This study demonstrated for the first time that CypA mediated the harmful effects of pericytes on BBB disruption after SAH, which potentially mediated by CD147/NF-κB/MMP9 signal, and junction protein degradation in the brain. By targeting CypA and pericytes, this study may provide new insights on the management of SAH patients.
Collapse
|
26
|
Liu Q, Yang Y, Fan X. Microvascular pericytes in brain-associated vascular disease. Biomed Pharmacother 2020; 121:109633. [DOI: 10.1016/j.biopha.2019.109633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 01/01/2023] Open
|
27
|
Gomez-Zepeda D, Taghi M, Scherrmann JM, Decleves X, Menet MC. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019; 12:pharmaceutics12010020. [PMID: 31878061 PMCID: PMC7022905 DOI: 10.3390/pharmaceutics12010020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Drug delivery into the brain is regulated by the blood-brain interfaces. The blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the blood-arachnoid barrier (BAB) regulate the exchange of substances between the blood and brain parenchyma. These selective barriers present a high impermeability to most substances, with the selective transport of nutrients and transporters preventing the entry and accumulation of possibly toxic molecules, comprising many therapeutic drugs. Transporters of the ATP-binding cassette (ABC) superfamily have an important role in drug delivery, because they extrude a broad molecular diversity of xenobiotics, including several anticancer drugs, preventing their entry into the brain. Gliomas are the most common primary tumors diagnosed in adults, which are often characterized by a poor prognosis, notably in the case of high-grade gliomas. Therapeutic treatments frequently fail due to the difficulty of delivering drugs through the brain barriers, adding to diverse mechanisms developed by the cancer, including the overexpression or expression de novo of ABC transporters in tumoral cells and/or in the endothelial cells forming the blood-brain tumor barrier (BBTB). Many models have been developed to study the phenotype, molecular characteristics, and function of the blood-brain interfaces as well as to evaluate drug permeability into the brain. These include in vitro, in vivo, and in silico models, which together can help us to better understand their implication in drug resistance and to develop new therapeutics or delivery strategies to improve the treatment of pathologies of the central nervous system (CNS). In this review, we present the principal characteristics of the blood-brain interfaces; then, we focus on the ABC transporters present on them and their implication in drug delivery; next, we present some of the most important models used for the study of drug transport; finally, we summarize the implication of ABC transporters in glioma and the BBTB in drug resistance and the strategies to improve the delivery of CNS anticancer drugs.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| | - Méryam Taghi
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Xavier Decleves
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Biologie du médicament et toxicologie, Hôpital Cochin, AP HP, 75006 Paris, France
| | - Marie-Claude Menet
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Hormonologie adulte, Hôpital Cochin, AP HP, 75006 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| |
Collapse
|
28
|
Su X, Huang L, Qu Y, Xiao D, Mu D. Pericytes in Cerebrovascular Diseases: An Emerging Therapeutic Target. Front Cell Neurosci 2019; 13:519. [PMID: 31824267 PMCID: PMC6882740 DOI: 10.3389/fncel.2019.00519] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022] Open
Abstract
Pericytes are functional components of the neurovascular unit (NVU) that are located around the blood vessels, and their roles in the regulation of cerebral health and diseases has been reported. Currently, the potential properties of pericytes as emerging therapeutic targets for cerebrovascular diseases have attracted considerable attention. Nonetheless, few reviews have comprehensively discussed pericytes and their roles in cerebrovascular diseases. Therefore, in this review, we not only summarized and described the basic characteristics of pericytes but also focused on clarifying the new understanding about the roles of pericytes in the pathogenesis of cerebrovascular diseases, including white matter injury (WMI), hypoxic-ischemic brain damage, depression, neovascular insufficiency disease, and Alzheimer's disease (AD). Furthermore, we summarized the current therapeutic strategies targeting pericytes for cerebrovascular diseases. Collectively, this review is aimed at providing a comprehensive understanding of pericytes and new insights about the use of pericytes as novel therapeutic targets for cerebrovascular diseases.
Collapse
Affiliation(s)
- Xiaojuan Su
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Lingyi Huang
- West China College of Stomatology, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dongqiong Xiao
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
29
|
Abdul Razzak R, Florence GJ, Gunn-Moore FJ. Approaches to CNS Drug Delivery with a Focus on Transporter-Mediated Transcytosis. Int J Mol Sci 2019; 20:E3108. [PMID: 31242683 PMCID: PMC6627589 DOI: 10.3390/ijms20123108] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/15/2019] [Accepted: 06/16/2019] [Indexed: 12/13/2022] Open
Abstract
Drug delivery to the central nervous system (CNS) conferred by brain barriers is a major obstacle in the development of effective neurotherapeutics. In this review, a classification of current approaches of clinical or investigational importance for the delivery of therapeutics to the CNS is presented. This classification includes the use of formulations administered systemically that can elicit transcytosis-mediated transport by interacting with transporters expressed by transvascular endothelial cells. Neurotherapeutics can also be delivered to the CNS by means of surgical intervention using specialized catheters or implantable reservoirs. Strategies for delivering drugs to the CNS have evolved tremendously during the last two decades, yet, some factors can affect the quality of data generated in preclinical investigation, which can hamper the extension of the applications of these strategies into clinically useful tools. Here, we disclose some of these factors and propose some solutions that may prove valuable at bridging the gap between preclinical findings and clinical trials.
Collapse
Affiliation(s)
- Rana Abdul Razzak
- Medical and Biological Sciences Building, School of Biology, University of St Andrews, St Andrews KY16 9TF, UK.
- Biomedical Science Research Centre, Schools of Chemistry and Biology, University of St Andrews, St Andrews KY16 9TF, UK.
| | - Gordon J Florence
- Biomedical Science Research Centre, Schools of Chemistry and Biology, University of St Andrews, St Andrews KY16 9TF, UK.
| | - Frank J Gunn-Moore
- Medical and Biological Sciences Building, School of Biology, University of St Andrews, St Andrews KY16 9TF, UK.
- Biomedical Science Research Centre, Schools of Chemistry and Biology, University of St Andrews, St Andrews KY16 9TF, UK.
| |
Collapse
|
30
|
|
31
|
Barreto RSN, Romagnolli P, Cereta AD, Coimbra-Campos LMC, Birbrair A, Miglino MA. Pericytes in the Placenta: Role in Placental Development and Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:125-151. [PMID: 30937867 DOI: 10.1007/978-3-030-11093-2_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The placenta is the most variable organ, in terms of structure, among the species. Besides it, all placental types have the same function: production of viable offspring, independent of pregnancy length, litter number, or invasion level. The angiogenesis is a central mechanism for placental functionality, due to proper maternal-fetal communication and exchanges. Much is known about the vasculature structure, but little is known about vasculature development and cellular interactions. Pericytes are perivascular cells that were described to control vasculature stability and permeability. Nowadays there are several new functions discovered, such as lymphocyte modulation and activation, macrophage-like phagocytic properties, tissue regenerative and repair processes, and also the ability to modulate stem cells, majorly the hematopoietic. In parallel, placental tissues are known to be a particularly immune microenvironment and a rich stem cell niche. The pericyte function plethora could be similar in the placental microenvironment and could have a central role in placental development and homeostasis.
Collapse
Affiliation(s)
- Rodrigo S N Barreto
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Patricia Romagnolli
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Andressa Daronco Cereta
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Leda M C Coimbra-Campos
- Department of Pathology, Federal University of Minas Gerais, Pampulha, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.,Department of Pathology, Federal University of Minas Gerais, Pampulha, Belo Horizonte, Brazil
| | - Maria Angelica Miglino
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil.
| |
Collapse
|
32
|
Shimizu F, Nishihara H, Kanda T. Blood-brain barrier dysfunction in immuno-mediated neurological diseases. Immunol Med 2018; 41:120-128. [PMID: 30938273 DOI: 10.1080/25785826.2018.1531190] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The blood-brain barrier (BBB) is the brain-specific endothelial cell barrier that is important for maintaining brain homeostasis and preventing the entry of toxic substances. Pathological BBB dysfunction is a critical step of the disease process in several immuno-mediated neurological diseases, including multiple sclerosis (MS), neuromyelitis optica (NMO), neuropsychiatric systemic lupus erythematosus (NPSLE) and neuro-Behçet diseases. The pathological findings from patients with secondary progressive (SP) MS, NMO and NPSLE showed leaky BBB in the active lesions. NMO is a disease with strong evidence of disease-specific and pathogenic autoantibodies (aquaporin 4 [AQP4] autoantibodies). In the development of NMO, circulating AQP4 autoantibodies need to pass through the BBB in order to reach AQP4 on the astrocyte endfeet. Strong evidence suggests that NPSLE is associated with the disruption of the BBB and NPSLE patients frequently have antibodies bound to endothelial cells in their sera. We recently identified two BBB-reactive autoantibodies in immuno-mediated neurological diseases: galectin-3 autoantibodies in SPMS and GRP78 autoantibodies in NMO. In the present review article, we describe the basic structure and cellular biology of the BBB, discuss recent insights regarding the pathophysiology of the BBB breakdown in the setting of immuno-mediated neurological diseases, and describe our recent findings of autoantibody-mediated BBB breakdown.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- a Department of Neurology and Clinical Neuroscience , Yamaguchi University Graduate School of Medicine , Ube , Japan
| | - Hideaki Nishihara
- a Department of Neurology and Clinical Neuroscience , Yamaguchi University Graduate School of Medicine , Ube , Japan
| | - Takashi Kanda
- a Department of Neurology and Clinical Neuroscience , Yamaguchi University Graduate School of Medicine , Ube , Japan
| |
Collapse
|
33
|
Liu Q, Wang X, Yi S. Pathophysiological Changes of Physical Barriers of Peripheral Nerves After Injury. Front Neurosci 2018; 12:597. [PMID: 30210280 PMCID: PMC6119778 DOI: 10.3389/fnins.2018.00597] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Peripheral nerves are composed of complex layered anatomical structures, including epineurium, perineurium, and endoneurium. Perineurium and endoneurium contain many physical barriers, including the blood-nerve barrier at endoneurial vessels and the perineurial barrier. These physical barriers help to eliminate flux penetration and thus contribute to the establishment of a stable microenvironment. In the current review, we introduce the anatomical compartments and physical barriers of peripheral nerves and then describe the cellular and molecular basis of peripheral physical barriers. We also specifically explore peripheral nerve injury-induced changes of peripheral physical barriers, including elevated endoneurial fluid pressure, increased leakage of tracer, decreased barrier-type endothelial cell ratio, and altered distributions and expressions of cellular junctional proteins. The understanding of the pathophysiological changes of physical barriers following peripheral nerve injury may provide a clue for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Qianyan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
34
|
Prazeres PHDM, Turquetti AOM, Azevedo PO, Barreto RSN, Miglino MA, Mintz A, Delbono O, Birbrair A. Perivascular cell αv integrins as a target to treat skeletal muscle fibrosis. Int J Biochem Cell Biol 2018; 99:109-113. [PMID: 29627438 PMCID: PMC6159891 DOI: 10.1016/j.biocel.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
Fibrosis following injury leads to aberrant regeneration and incomplete functional recovery of skeletal muscle, but the lack of detailed knowledge about the cellular and molecular mechanisms involved hampers the design of effective treatments. Using state-of-the-art technologies, Murray et al. (2017) found that perivascular PDGFRβ-expressing cells generate fibrotic cells in the skeletal muscle. Strikingly, genetic deletion of αv integrins from perivascular PDGFRβ-expressing cells significantly inhibited skeletal muscle fibrosis without affecting muscle vascularization or regeneration. In addition, the authors showed that a small molecule inhibitor of αv integrins, CWHM 12, attenuates skeletal muscle fibrosis. From a drug-development perspective, this study identifies a new cellular and molecular target to treat skeletal muscle fibrosis.
Collapse
Affiliation(s)
- Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anaelise O M Turquetti
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo S N Barreto
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Maria A Miglino
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
35
|
Azevedo PO, Sena IFG, Andreotti JP, Carvalho-Tavares J, Alves-Filho JC, Cunha TM, Cunha FQ, Mintz A, Birbrair A. Pericytes modulate myelination in the central nervous system. J Cell Physiol 2018; 233:5523-5529. [PMID: 29215724 DOI: 10.1002/jcp.26348] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis is a highly prevalent chronic demyelinating disease of the central nervous system. Remyelination is the major therapeutic goal for this disorder. The lack of detailed knowledge about the cellular and molecular mechanisms involved in myelination restricts the design of effective treatments. A recent study by using [De La Fuente et al. (2017) Cell Reports, 20(8): 1755-1764] by using state-of-the-art techniques, including pericyte-deficient mice in combination with induced demyelination, reveal that pericytes participate in central nervous system regeneration. Strikingly, pericytes presence is essential for oligodendrocyte progenitors differentiation and myelin formation during remyelination in the brain. The emerging knowledge from this research will be important for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - Juliana Carvalho-Tavares
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil
| | - José C Alves-Filho
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, New York
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerias, Brazil.,Department of Radiology, Columbia University Medical Center, New York, New York
| |
Collapse
|
36
|
Castro V, Skowronska M, Lombardi J, He J, Seth N, Velichkovska M, Toborek M. Occludin regulates glucose uptake and ATP production in pericytes by influencing AMP-activated protein kinase activity. J Cereb Blood Flow Metab 2018; 38:317-332. [PMID: 28718701 PMCID: PMC5951017 DOI: 10.1177/0271678x17720816] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Energetic regulation at the blood-brain barrier is critical for maintaining its integrity, transport capabilities, and brain demands for glucose. However, the underlying mechanisms that regulate these processes are still poorly explored. We recently characterized the protein occludin as a NADH oxidase and demonstrated its influence on the expression and activation of the histone deacetylase SIRT-1. Because SIRT-1 works in concert with AMP-activated protein kinase (AMPK) (AMPK), we investigated the impact of occludin on this metabolic switch. Here we show that in blood-brain barrier pericytes, occludin promotes AMPK expression and activation, influencing the expression of glucose transporters GLUT-1 and GLUT-4, glucose uptake, and ATP content. Furthermore, occludin expression, AMP-dependent protein kinase activity, and glucose uptake were altered under inflammatory (TNFα) and infectious (HIV) conditions. We also show that pericytes share glucose and mitochondria with astrocytes, and that occludin levels modify the ability of pericytes to share those energetic resources. In addition, we demonstrate that murine mitochondria can be transferred from live brain microvessels to energetically impaired human astrocytes, promoting their survival. Our findings demonstrate that occludin plays an important role in blood-brain barrier pericyte metabolism by influencing AMPK protein kinase activity, glucose uptake, ATP production, and by regulating the ability of pericytes to interact metabolically with astrocytes.
Collapse
Affiliation(s)
- Victor Castro
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Marta Skowronska
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jorge Lombardi
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jane He
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Neil Seth
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Martina Velichkovska
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
37
|
Pereira CD, Martins F, Wiltfang J, da Cruz e Silva OA, Rebelo S. ABC Transporters Are Key Players in Alzheimer’s Disease. J Alzheimers Dis 2017; 61:463-485. [DOI: 10.3233/jad-170639] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Cátia D. Pereira
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Filipa Martins
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Jens Wiltfang
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Odete A.B. da Cruz e Silva
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Sandra Rebelo
- Department of Medical Sciences, Neuroscience and Signalling Laboratory, Institute for Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
38
|
Neural Vascular Mechanism for the Cerebral Blood Flow Autoregulation after Hemorrhagic Stroke. Neural Plast 2017; 2017:5819514. [PMID: 29104807 PMCID: PMC5634612 DOI: 10.1155/2017/5819514] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/11/2017] [Indexed: 12/21/2022] Open
Abstract
During the initial stages of hemorrhagic stroke, including intracerebral hemorrhage and subarachnoid hemorrhage, the reflex mechanisms are activated to protect cerebral perfusion, but secondary dysfunction of cerebral flow autoregulation will eventually reduce global cerebral blood flow and the delivery of metabolic substrates, leading to generalized cerebral ischemia, hypoxia, and ultimately, neuronal cell death. Cerebral blood flow is controlled by various regulatory mechanisms, including prevailing arterial pressure, intracranial pressure, arterial blood gases, neural activity, and metabolic demand. Evoked by the concept of vascular neural network, the unveiled neural vascular mechanism gains more and more attentions. Astrocyte, neuron, pericyte, endothelium, and so forth are formed as a communicate network to regulate with each other as well as the cerebral blood flow. However, the signaling molecules responsible for this communication between these new players and blood vessels are yet to be definitively confirmed. Recent evidence suggested the pivotal role of transcriptional mechanism, including but not limited to miRNA, lncRNA, exosome, and so forth, for the cerebral blood flow autoregulation. In the present review, we sought to summarize the hemodynamic changes and underline neural vascular mechanism for cerebral blood flow autoregulation in stroke-prone state and after hemorrhagic stroke and hopefully provide more systematic and innovative research interests for the pathophysiology and therapeutic strategies of hemorrhagic stroke.
Collapse
|
39
|
BCRP expression in schwannoma, plexiform neurofibroma and MPNST. Oncotarget 2017; 8:88751-88759. [PMID: 29179472 PMCID: PMC5687642 DOI: 10.18632/oncotarget.21075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/17/2017] [Indexed: 01/10/2023] Open
Abstract
Background peripheral nerve sheath tumors comprise a broad spectrum of neoplasms. Vestibular schwannomas and plexiform neurofibromas are symptomatic albeit benign, but a subset of the latter pre-malignant lesions will transform to malignant peripheral nerve sheath tumors (MPNST). Surgery and radiotherapy are the primary strategies to treat these tumors. Intrinsic resistance to drug therapy characterizes all three tumor subtypes. The breast cancer resistance protein BCRP is a transmembrane efflux transporter considered to play a key role in various biological barriers such as the blood brain barrier. At the same time it is associated with drug resistance in various tumors. Its potential role in drug resistant tumors of the peripheral nervous system is largely unknown. Objective to assess if BCRP is expressed in vestibular schwannomas, plexiform neurofibromas and MPNST. Material and methods immunohistochemical staining for BCRP was performed on a tissue microarray composed out of 22 vestibular schwannomas, 10 plexiform neurofibromas and 18 MPNSTs. Results sixteen out of twenty-two vestibular schwannomas (73%), nine out of ten plexiform neurofibromas (90%) and six out of eighteen MPNST (33%) expressed BCRP in the vasculature. Tumor cells were negative. Conclusion BCRP is present in the vasculature of vestibular schwannomas, plexiform neurofibromas and MPSNT. Therefore, it may reduce the drug exposure of underlying tumor tissues and potentially cause failure of drug therapy.
Collapse
|
40
|
The role of non-endothelial cells on the penetration of nanoparticles through the blood brain barrier. Prog Neurobiol 2017; 159:39-49. [PMID: 28899762 DOI: 10.1016/j.pneurobio.2017.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/30/2017] [Accepted: 09/08/2017] [Indexed: 12/26/2022]
Abstract
The blood brain barrier (BBB) is a well-established cell-based membrane that circumvents the central nervous system (CNS), protecting it from harmful substances. Due to its robustness and cell integrity, it is also an outstanding opponent when it comes to the delivery of several therapeutic agents to the brain, which requires the crossing through its highly-organized structure. This regulation and cell-cell communications occur mostly between astrocytes, pericytes and endothelial cells. Therefore, alternative ways to deliver drugs to the CNS, overcoming the BBB are required, to improve the efficacy of brain target drugs. Nanoparticles emerge here as a promising drug delivery strategy, due to their ability of high drug loading and the capability to exploit specific delivery pathways that most drugs are unable to when administered freely, increasing their bioavailability in the CNS. Thus, further attempts to assess the possible influence of non-endothelial may have on the BBB translocation of nanoparticles are here revised. Furthermore, the use of macrophages and/or monocytes as nanoparticle delivery cells are also approached. Lastly, the temporarily disruption of the overall organization and normal structure of the BBB to promote the penetration of nanoparticles aimed at the CNS is described, as a synergistic path.
Collapse
|
41
|
Takahashi S, Maeda T, Sano Y, Nishihara H, Takeshita Y, Shimizu F, Kanda T. Active form of vitamin D directly protects the blood-brain barrier in multiple sclerosis. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/cen3.12398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Shiori Takahashi
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | - Toshihiko Maeda
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | - Hideaki Nishihara
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | - Yukio Takeshita
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| |
Collapse
|
42
|
Birbrair A, Borges IDT, Gilson Sena IF, Almeida GG, da Silva Meirelles L, Gonçalves R, Mintz A, Delbono O. How Plastic Are Pericytes? Stem Cells Dev 2017; 26:1013-1019. [PMID: 28490256 PMCID: PMC5512298 DOI: 10.1089/scd.2017.0044] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/27/2017] [Indexed: 01/18/2023] Open
Abstract
Pericytes are defined by both their anatomical location and molecular markers. Numerous publications have reported their role as stem cells, contributing to the formation of tissues other than blood vessels. However, using cell-lineage tracing in a new transgenic mouse model, a recent study shows that in the context of aging and some pathologies, Tbx18+ pericytes do not function as stem cells in vivo. This study challenges the current view that pericytes can differentiate into other cells and reopen questions about their plasticity. This emerging knowledge is important not only for our understanding of development but may also inform treatments for diseases.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York
| | | | | | | | | | - Ricardo Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
43
|
Dias Moura Prazeres PH, Sena IFG, Borges IDT, de Azevedo PO, Andreotti JP, de Paiva AE, de Almeida VM, de Paula Guerra DA, Pinheiro Dos Santos GS, Mintz A, Delbono O, Birbrair A. Pericytes are heterogeneous in their origin within the same tissue. Dev Biol 2017; 427:6-11. [PMID: 28479340 PMCID: PMC6076854 DOI: 10.1016/j.ydbio.2017.05.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022]
Abstract
Pericytes heterogeneity is based on their morphology, distribution, and markers. It is well known that pericytes from different organs may have distinct embryonic sources. Yamazaki et al. (2017) using several transgenic mouse model reveal by cell-lineage tracing that pericytes are even more heterogeneous than previously appreciated. This study shows that pericytes from within the same tissue may be heterogeneous in their origin. Remarkably, a subpopulation of embryonic dermal pericytes derives from the hematopoietic lineage, an unexpected source. Reconstructing the lineage of pericytes is central to understanding development, and also for the diagnosis and treatment of diseases in which pericytes play important roles.
Collapse
Affiliation(s)
| | | | | | | | - Julia Peres Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Emília de Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA; Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
44
|
Pericytes: The Role of Multipotent Stem Cells in Vascular Maintenance and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:69-86. [PMID: 29282647 DOI: 10.1007/5584_2017_138] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Blood vessels consist of an inner endothelial cell layer lining the vessel wall and perivascular pericytes, also known as mural cells, which envelop the vascular tube surface. Pericytes have recently been recognized for their central role in blood vessel formation. Pericytes are multipotent cells that are heterogeneous in their origin, function, morphology and surface markers. Similar to other types of stem cells, pericytes act as a repair system in response to injury by maintaining the structural integrity of blood vessels. Several studies have shown that blood vessels lacking pericytes become hyperdilated and haemorrhagic, leading to vascular complications ranging from diabetic retinopathy to embryonic death. The role of pericytes is not restricted to the formation and development of the vasculature: they have been shown to possess stem cell-like characteristics and may differentiate into cell types from different lineages. Recent discoveries regarding the contribution of pericytes to tumour metastasis and the maintenance of tumour vascular supply and angiogenesis have led researchers to propose targeting pericytes with anti-angiogenic therapies. In this review, we will examine the different physiological roles of pericytes, their differentiation potential, and how they interact with surrounding cells to ensure the integrity of blood vessel formation and maintenance.
Collapse
|
45
|
Castro V, Bertrand L, Luethen M, Dabrowski S, Lombardi J, Morgan L, Sharova N, Stevenson M, Blasig IE, Toborek M. Occludin controls HIV transcription in brain pericytes via regulation of SIRT-1 activation. FASEB J 2015; 30:1234-46. [PMID: 26601824 DOI: 10.1096/fj.15-277673] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/16/2015] [Indexed: 12/22/2022]
Abstract
HIV invades the brain early after infection; however, its interactions with the cells of the blood-brain barrier (BBB) remain poorly understood. Our goal was to evaluate the role of occludin, one of the tight junction proteins that regulate BBB functions in HIV infection of BBB pericytes. We provide evidence that occludin levels largely control the metabolic responses of human pericytes to HIV. Occludin in BBB pericytes decreased by 10% during the first 48 h after HIV infection, correlating with increased nuclear translocation of the gene repressor C-terminal-binding protein (CtBP)-1 and NFκB-p65 activation. These changes were associated with decreased expression and activation of the class III histone deacetylase sirtuin (SIRT)-1. Occludin levels recovered 96 h after infection, restoring SIRT-1 and reducing HIV transcription to 20% of its highest values. We characterized occludin biochemically as a novel NADH oxidase that controls the expression and activation of SIRT-1. The inverse correlation between occludin and HIV transcription was then replicated in human primary macrophages and differentiated monocytic U937 cells, in which occludin silencing resulted in 75 and 250% increased viral transcription, respectively. Our work shows that occludin has previously unsuspected metabolic properties and is a target of HIV infection, opening the possibility of designing novel pharmacological approaches to control HIV transcription.
Collapse
Affiliation(s)
- Victor Castro
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Luc Bertrand
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Mareen Luethen
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Sebastian Dabrowski
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Jorge Lombardi
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Laura Morgan
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Natalia Sharova
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Mario Stevenson
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Ingolf E Blasig
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Michal Toborek
- *Department of Biochemistry and Molecular Biology and Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Biology, Freie Universität Berlin, Berlin, Germany; and Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| |
Collapse
|
46
|
Capek S, Howe BM, Amrami KK, Spinner RJ. Perineural spread of pelvic malignancies to the lumbosacral plexus and beyond: clinical and imaging patterns. Neurosurg Focus 2015; 39:E14. [DOI: 10.3171/2015.7.focus15209] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT
Perineural spread along pelvic autonomie nerves has emerged as a logical, anatomical explanation for selected cases of neoplastic lumbosacral plexopathy (LSP) in patients with prostate, bladder, rectal, and cervical cancer. The authors wondered whether common radiological and clinical patterns shared by various types of pelvic cancer exist.
METHODS
The authors retrospectively reviewed their institutional series of 17 cases concluded as perineural tumor spread. All available history, physical examination, electrodiagnostic studies, biopsy data and imaging studies, evidence of other metastatic disease, and follow-up were recorded in detail. The series was divided into 2 groups: cases with neoplastic lumbosacral plexopathy confirmed by biopsy (Group A) and cases included based on imaging characteristics despite the lack of biopsy or negative biopsy results (Group B).
RESULTS
Group A comprised 10 patients (mean age 69 years); 9 patients were symptomatic and 1 was asymptomatic. The L5–S1 spinal nerves and sciatic nerve were most frequently involved. Three patients had intradural extension. Seven patients were alive at last follow-up. Group B consisted of 7 patients (mean age 64 years); 4 patients were symptomatic, 2 were asymptomatic, and 1 had only imaging available. The L5–S1 spinal nerves and the sciatic nerve were most frequently involved. No patients had intradural extension. Four patients were alive at last follow-up.
CONCLUSIONS
The authors provide a unifying theory to explain lumbosacral plexopathy in select cases of various pelvic neoplasms. The tumor cells can use splanchnic nerves as conduits and spread from the end organ to the lumbosacral plexus. Tumor can continue to spread along osseous and muscle nerve branches, resulting in muscle and bone “metastases.” Radiological studies show a reproducible, although nonspecific pattern, and the same applies to clinical presentation.
Collapse
Affiliation(s)
- Stepan Capek
- Departments of 1Neurosurgery and
- 22nd Faculty of Medicine, Charles University in Prague, Czech Republic
| | | | | | | |
Collapse
|
47
|
Neuhaus W, Schlundt M, Fehrholz M, Ehrke A, Kunzmann S, Liebner S, Speer CP, Förster CY. Multiple Antenatal Dexamethasone Treatment Alters Brain Vessel Differentiation in Newborn Mouse Pups. PLoS One 2015; 10:e0136221. [PMID: 26274818 PMCID: PMC4537167 DOI: 10.1371/journal.pone.0136221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022] Open
Abstract
Antenatal steroid treatment decreases morbidity and mortality in premature infants through the maturation of lung tissue, which enables sufficient breathing performance. However, clinical and animal studies have shown that repeated doses of glucocorticoids such as dexamethasone and betamethasone lead to long-term adverse effects on brain development. Therefore, we established a mouse model for antenatal dexamethasone treatment to investigate the effects of dexamethasone on brain vessel differentiation towards the blood-brain barrier (BBB) phenotype, focusing on molecular marker analysis. The major findings were that in total brains on postnatal day (PN) 4 triple antenatal dexamethasone treatment significantly downregulated the tight junction protein claudin-5, the endothelial marker Pecam-1/CD31, the glucocorticoid receptor, the NR1 subunit of the N-methyl-D-aspartate receptor, and Abc transporters (Abcb1a, Abcg2 Abcc4). Less pronounced effects were found after single antenatal dexamethasone treatment and in PN10 samples. Comparisons of total brain samples with isolated brain endothelial cells together with the stainings for Pecam-1/CD31 and claudin-5 led to the assumption that the morphology of brain vessels is affected by antenatal dexamethasone treatment at PN4. On the mRNA level markers for angiogenesis, the sonic hedgehog and the Wnt pathway were downregulated in PN4 samples, suggesting fundamental changes in brain vascularization and/or differentiation. In conclusion, we provided a first comprehensive molecular basis for the adverse effects of multiple antenatal dexamethasone treatment on brain vessel differentiation.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Department of Anaesthesia and Critical Care, University of Wuerzburg, Wuerzburg, Germany
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
- * E-mail: (WN); (CF)
| | - Marian Schlundt
- Department of Anaesthesia and Critical Care, University of Wuerzburg, Wuerzburg, Germany
| | - Markus Fehrholz
- University Children’s Hospital Wuerzburg, Wuerzburg, Germany
| | - Alexander Ehrke
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | | | - Stefan Liebner
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | | | - Carola Y. Förster
- Department of Anaesthesia and Critical Care, University of Wuerzburg, Wuerzburg, Germany
- * E-mail: (WN); (CF)
| |
Collapse
|
48
|
The evolving roles of pericyte in early brain injury after subarachnoid hemorrhage. Brain Res 2015; 1623:110-22. [PMID: 25982598 DOI: 10.1016/j.brainres.2015.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/02/2015] [Accepted: 05/04/2015] [Indexed: 12/21/2022]
Abstract
Despite accumulated understanding on the mechanisms of early brain injury and improved management of subarachnoid hemorrhage (SAH), it is still one of the serious and refractory health problems around the world. Traditionally, pericyte, served as capillary contraction handler, is recently considered as the main participant of microcirculation regulation in SAH pathophysiology. However, accumulate evidences indicate that pericyte is much more than we already know. Therefore, we briefly review the characteristics, regulation pathways and functions of pericyte, aim to summarize the evolving new pathophysiological roles of pericyte that are implicated in early brain injury after SAH and to improve our understanding in order to explore potential novel therapeutic options for patients with SAH. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
|
49
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci (Lond) 2015; 128:81-93. [PMID: 25236972 PMCID: PMC4200531 DOI: 10.1042/cs20140278] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Perivascular multipotent cells, pericytes, contribute to the generation and repair of various tissues in response to injury. They are heterogeneous in their morphology, distribution, origin and markers, and elucidating their molecular and cellular differences may inform novel treatments for disorders in which tissue regeneration is either impaired or excessive. Moreover, these discoveries offer novel cellular targets for therapeutic approaches to many diseases. This review discusses recent studies that support the concept that pericyte subtypes play a distinctive role in myogenesis, neurogenesis, adipogenesis, fibrogenesis and angiogenesis.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| |
Collapse
|
50
|
Immortalized multipotent pericytes derived from the vasa vasorum in the injured vasculature. A cellular tool for studies of vascular remodeling and regeneration. J Transl Med 2014; 94:1340-54. [PMID: 25329003 DOI: 10.1038/labinvest.2014.121] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 07/14/2014] [Accepted: 07/27/2014] [Indexed: 01/09/2023] Open
Abstract
Adventitial microvessels, vasa vasorum in the vessel walls, have an active role in the vascular remodeling, although its mechanisms are still unclear. It has been reported that microvascular pericytes (PCs) possess mesenchymal plasticity. Therefore, microvessels would serve as a systemic reservoir of stem cells and contribute to the tissues remodeling. However, most aspects of the biology of multipotent PCs (mPCs), in particular of pathological microvessels are still obscure because of the lack of appropriate methods to detect and isolate these cells. In order to examine the characteristics of mPCs, we established immortalized cells residing in adventitial capillary growing at the injured vascular walls. We recently developed in vivo angiogenesis to observe adventitial microvessels using collagen-coated tube (CCT), which also can be used as an adventitial microvessel-rich tissue. By using the CCT, CD146- or NG2-positive cells were isolated from the adventitial microvessels in the injured arteries of mice harboring a temperature-sensitive SV40 T-antigen gene. Several capillary-derived endothelial cells (cECs) and PCs (cPCs) cell lines were established. cECs and cPCs maintain a number of key endothelial and PC features. Co-incubation of cPCs with cECs formed capillary-like structure in Matrigel. Three out of six cPC lines, termed capillary mPCs demonstrated both mesenchymal stem cell- and neuronal stem cell-like phenotypes, differentiating effectively into adipocytes, osteoblasts, as well as schwann cells. mPCs differentiated to ECs and PCs, and formed capillary-like structure on their own. Transplanted DsRed-expressing mPCs were resident in the capillary and muscle fibers and promoted angiogenesis and myogenesis in damaged skeletal muscle. Adventitial mPCs possess transdifferentiation potential with unique phenotypes, including the reconstitution of capillary-like structures. Their phenotype would contribute to the pathological angiogenesis associated with vascular remodeling. These cell lines also provide a reproducible cellular tool for high-throughput studies on angiogenesis, vascular remodeling, and regeneration as well.
Collapse
|