1
|
Zheng T, Jiang T, Ma H, Zhu Y, Wang M. Targeting PI3K/Akt in Cerebral Ischemia Reperfusion Injury Alleviation: From Signaling Networks to Targeted Therapy. Mol Neurobiol 2024; 61:7930-7949. [PMID: 38441860 DOI: 10.1007/s12035-024-04039-1] [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: 11/01/2023] [Accepted: 02/09/2024] [Indexed: 09/21/2024]
Abstract
Ischemia/reperfusion (I/R) injury is a pathological event that results in reperfusion due to low blood flow to an organ. Cerebral ischemia is a common cerebrovascular disease with high mortality, and reperfusion is the current standard intervention. However, reperfusion may further induce cellular damage and dysfunction known as cerebral ischemia/reperfusion injury (CIRI). Currently, strategies for the clinical management of CIRI are limited, necessitating the exploration of novel and efficacious treatment modalities for the benefit of patients. PI3K/Akt signaling pathway is an important cellular process associated with the disease. Stimulation of the PI3K/Akt pathway enhances I/R injury in multiple organs such as heart, brain, lung, and liver. It stands as a pivotal signaling pathway crucial for diminishing cerebral infarction size and safeguarding the functionality of brain tissue after CIRI. During CIRI, activation of the PI3K/Akt pathway exhibits a protective effect on CIRI. Furthermore, activation of the PI3K/Akt pathway has the potential to augment the activity of antioxidant enzymes, resulting in a decrease in reactive oxygen species (ROS) and the associated oxidative stress. Meanwhile, PI3K/Akt plays a neuroprotective role by inhibiting inflammatory responses and apoptosis. For example, PI3K/Akt interacts with NF-κB, Nrf2, and MAPK signaling pathways to mitigate CIRI. This article is aimed to explore the pivotal role and underlying mechanism of PI3K/Akt in ameliorating CIRI and investigate the influence of ischemic preconditioning and post-processing, as well as the impact of pertinent drugs or activators targeting the PI3K/Akt pathway on CIRI. The primary objective is to furnish compelling evidence supporting the activation of PI3K/Akt in the context of CIRI, elucidating its mechanistic intricacies. By doing so, the paper aims to underscore the critical contribution of PI3K/Akt in mitigating CIRI, providing a theoretical foundation for considering the PI3K/Akt pathway as a viable target for CIRI treatment.
Collapse
Affiliation(s)
- Ting Zheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Taotao Jiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Hongxiang Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Yanping Zhu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Manxia Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
2
|
Cui D, Yamamoto K, Ikeda E. High-Mannose-Type Glycan of Basigin in Endothelial Cells Is Essential for the Opening of the Blood-Brain Barrier Induced by Hypoxia, Cyclophilin A, or Tumor Necrosis Factor-α. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:612-625. [PMID: 38040091 DOI: 10.1016/j.ajpath.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023]
Abstract
Pathologic opening of the blood-brain barrier accelerates the progression of various neural diseases. Basigin, as an essential molecule for the opening of the blood-brain barrier, is a highly glycosylated transmembrane molecule specified in barrier-forming endothelial cells. This study analyzed the involvement of basigin in the regulation of the blood-brain barrier focusing on its glycosylation forms. First, basigin was found to be expressed as cell surface molecules with complex-type glycan as well as those with high-mannose-type glycan in barrier-forming endothelial cells. Monolayers of endothelial cells with suppressed expression of basigin with high-mannose-type glycan were then prepared and exposed to pathologic stimuli. These monolayers retained their barrier-forming properties even in the presence of pathologic stimuli, although their expression of basigin with complex-type glycan was maintained. In vivo, the blood-brain barrier in mice pretreated intravenously with endoglycosidase H was protected from opening under pathologic stimuli. Pathologically opened blood-brain barrier in streptozotocin-injected mice was successfully closed by intravenous injection of endoglycosidase H. These results show that high-mannose-type glycan of the basigin molecule is essential for the opening of the blood-brain barrier and therefore a specific target for protection as well as restoration of pathologic opening of the blood-brain barrier.
Collapse
Affiliation(s)
- Dan Cui
- Department of Pathology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Kazuo Yamamoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Eiji Ikeda
- Department of Pathology, Yamaguchi University Graduate School of Medicine, Ube, Japan.
| |
Collapse
|
3
|
Yu N, Cui H, Jin S, Liu P, Fang Y, Sun F, Cao Y, Yuan B, Xie Y, Duan W, Ma C. IL-6 from cerebrospinal fluid causes widespread pain via STAT3-mediated astrocytosis in chronic constriction injury of the infraorbital nerve. J Neuroinflammation 2024; 21:60. [PMID: 38419042 PMCID: PMC10900663 DOI: 10.1186/s12974-024-03049-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND The spinal inflammatory signal often spreads to distant segments, accompanied by widespread pain symptom under neuropathological conditions. Multiple cytokines are released into the cerebrospinal fluid (CSF), potentially inducing the activation of an inflammatory cascade at remote segments through CSF flow. However, the detailed alteration of CSF in neuropathic pain and its specific role in widespread pain remain obscure. METHODS A chronic constriction injury of the infraorbital nerve (CCI-ION) model was constructed, and pain-related behavior was observed on the 7th, 14th, 21st, and 28th days post surgery, in both vibrissa pads and hind paws. CSF from CCI-ION rats was transplanted to naïve rats through intracisternal injection, and thermal and mechanical allodynia were measured in hind paws. The alteration of inflammatory cytokines in CCI-ION's CSF was detected using an antibody array and bioinformatic analysis. Pharmacological intervention targeting the changed cytokine in the CSF and downstream signaling was performed to evaluate its role in widespread pain. RESULTS CCI-ION induced local pain in vibrissa pads together with widespread pain in hind paws. CCI-ION's CSF transplantation, compared with sham CSF, contributed to vibrissa pad pain and hind paw pain in recipient rats. Among the measured cytokines, interleukin-6 (IL-6) and leptin were increased in CCI-ION's CSF, while interleukin-13 (IL-13) was significantly reduced. Furthermore, the concentration of CSF IL-6 was correlated with nerve injury extent, which gated the occurrence of widespread pain. Both astrocytes and microglia were increased in remote segments of the CCI-ION model, while the inhibition of astrocytes in remote segments, but not microglia, significantly alleviated widespread pain. Mechanically, astroglial signal transducer and activator of transcription 3 (STAT3) in remote segments were activated by CSF IL-6, the inhibition of which significantly mitigated widespread pain in CCI-ION. CONCLUSION IL-6 was induced in the CSF of the CCI-ION model, triggering widespread pain via activating astrocyte STAT3 signal in remote segments. Therapies targeting IL-6/STAT3 signaling might serve as a promising strategy for the widespread pain symptom under neuropathological conditions.
Collapse
Affiliation(s)
- Ning Yu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Huan Cui
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Sixuan Jin
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Penghao Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45# Changchun Street, Xicheng District, Beijing, 100053, China
- Lab of Spinal Cord Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Yehong Fang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengrun Sun
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Yan Cao
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Bo Yuan
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Yikuan Xie
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China
| | - Wanru Duan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45# Changchun Street, Xicheng District, Beijing, 100053, China.
- Lab of Spinal Cord Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China.
| | - Chao Ma
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Joint Laboratory of Anesthesia and Pain, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, No. 5 DongDanSanTiao, Dongcheng District, Beijing, 100005, China.
- National Human Brain Bank for Development and Function, Beijing, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
4
|
Kuo YC, Yen MH, De S, Rajesh R, Tai CK. Optimized lipopolymers with curcumin to enhance AZD5582 and GDC0152 activity and downregulate inhibitors of apoptosis proteins in glioblastoma multiforme. BIOMATERIALS ADVANCES 2023; 154:213639. [PMID: 37793310 DOI: 10.1016/j.bioadv.2023.213639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/07/2023] [Accepted: 09/24/2023] [Indexed: 10/06/2023]
Abstract
Inhibition to glioblastoma multiforme (GBM) propagation is a critical challenge in clinical practice because binding of inhibitors of apoptosis proteins (IAPs) to caspase prevents cancer cells from death. In this study, folic acid (FA), lactoferrin (Lf) and rabies virus glycoprotein (RVG) were grafted on lipopolymers (LPs) composed of poly(ε-caprolactone) and Compritol 888 ATO to encapsulate AZD5582 (AZD), GDC0152 (GDC) and curcumin (CURC). The standard deviations of initial particle diameter and particle diameter after storage for 30 days were involved in LP composition optimization. The functionalized LPs were used to permeate the blood-brain barrier (BBB) and constrain IAP quantity in GBM cells. Experimental results revealed that an increase in Span 20 (emulsifier) concentration enlarged the size of LPs, and enhanced the entrapment and releasing efficiency of AZD, DGC and CURC. 1H nuclear magnetic resonance spectra showed that the hydrogen bonds between the LPs and drugs supported the sustained release of AZD, DGC and CURC from the LPs. The LPs modified with the three targeting biomolecules facilitated the penetration of AZD, GDC and CURC across the BBB, and could recognize U87MG cells and human brain cancer stem cells. Immunofluorescence staining, flow cytometry and western blot demonstrated that CURC-incorporated LPs enhanced AZD and GDC activity in suppressing cellular IAP 1 (cIAP1) and X-linked IAP (XIAP) levels, and raising caspase-3 level in GBM. Surface FA, Lf and RVG also promoted the ability of the drug-loaded LPs to avoid carcinoma growth. The current FA-, Lf- and RVG-crosslinked LPs carrying AZD, DGC and CURC can be promising in hindering IAP expressions for GBM management.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC; Advanced Institute of Manufacturing with High-tech Innovations, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC.
| | - Meng-Hui Yen
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | - Sourav De
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | - Chien-Kuo Tai
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| |
Collapse
|
5
|
Chang K, Gao D, Yan J, Lin L, Cui T, Lu S. Critical Roles of Protein Arginine Methylation in the Central Nervous System. Mol Neurobiol 2023; 60:6060-6091. [PMID: 37415067 DOI: 10.1007/s12035-023-03465-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/24/2023] [Indexed: 07/08/2023]
Abstract
A remarkable post-transitional modification of both histones and non-histone proteins is arginine methylation. Methylation of arginine residues is crucial for a wide range of cellular process, including signal transduction, DNA repair, gene expression, mRNA splicing, and protein interaction. Arginine methylation is modulated by arginine methyltransferases and demethylases, like protein arginine methyltransferase (PRMTs) and Jumonji C (JmjC) domain containing (JMJD) proteins. Symmetric dimethylarginine and asymmetric dimethylarginine, metabolic products of the PRMTs and JMJD proteins, can be changed by abnormal expression of these proteins. Many pathologies including cancer, inflammation and immune responses have been closely linked to aberrant arginine methylation. Currently, the majority of the literature discusses the substrate specificity and function of arginine methylation in the pathogenesis and prognosis of cancers. Numerous investigations on the roles of arginine methylation in the central nervous system (CNS) have so far been conducted. In this review, we display the biochemistry of arginine methylation and provide an overview of the regulatory mechanism of arginine methyltransferases and demethylases. We also highlight physiological functions of arginine methylation in the CNS and the significance of arginine methylation in a variety of neurological diseases such as brain cancers, neurodegenerative diseases and neurodevelopmental disorders. Furthermore, we summarize PRMT inhibitors and molecular functions of arginine methylation. Finally, we pose important questions that require further research to comprehend the roles of arginine methylation in the CNS and discover more effective targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Kewei Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Dan Gao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jidong Yan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Liyan Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Tingting Cui
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Shemin Lu
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
- Department of Biochemistry and Molecular Biology, and Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
6
|
Mughis H, Lye P, Matthews SG, Bloise E. Hypoxia modifies levels of the SARS-CoV-2 cell entry proteins, angiotensin-converting enzyme 2, and furin in fetal human brain endothelial cells. Am J Obstet Gynecol MFM 2023; 5:101126. [PMID: 37562534 DOI: 10.1016/j.ajogmf.2023.101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND It is not known whether human fetal brain endothelial cells that form the blood-brain barrier express angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin, which are SARS-CoV-2 cell entry proteins. Moreover, it is unclear whether hypoxia, commonly observed during severe maternal COVID-19, can modify their level of expression. We hypothesized that human fetal brain endothelial cells isolated from early- and midpregnancy brain microvessels express angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin. Furthermore, we hypothesized that hypoxia modifies their expression levels in a gestational age- and time-of-exposure-dependent manner. OBJECTIVE This study aimed to investigate whether early- and midpregnancy human fetal brain endothelial cells express angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin SARS-CoV-2-associated cell entry proteins and to determine the effects of hypoxia on angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin expression levels in human fetal brain endothelial cells. STUDY DESIGN This was a prospective study where human fetal brain endothelial cells isolated from early-pregnancy (12.4±0.7 weeks of gestation) and midpregnancy (17.9±0.5 weeks of gestation) fetal brain microvessels (6 per group) were exposed to different oxygen tensions (20%, 5%, and 1% oxygen) for 6, 24, and 48 hours. Angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin messenger RNA and protein levels and localization were assessed using quantitative polymerase chain reaction, Western blot testing, and immunofluorescence. RESULTS Angiotensin-converting enzyme 2, transmembrane serine protease 2, and furin co-localize with the endothelial cell marker von Willebrand factor in human fetal brain endothelial cells isolated from early pregnancy and midpregnancy. In early pregnancy, TMPRSS2 messenger RNA expression was decreased by 5% oxygen compared with 20% oxygen after 6 hours of exposure (P<.05). In midpregnancy, 5% oxygen down-regulated ACE2 messenger RNA compared with 20% oxygen after 24 hours (P<.05). Furin messenger RNA expression was decreased under 5% and 1% oxygen compared with 20% oxygen (P<.05) after 24 hours. In midpregnancy, angiotensin-converting enzyme 2 protein levels were decreased under 5% and 1% oxygen (P<.001) after 24 hours. In contrast, furin protein levels were increased under 1% oxygen compared with 20% oxygen after 24 hours (P<.05). At 48 hours, 1% oxygen increased angiotensin-converting enzyme 2 protein levels compared with 20% oxygen (P<.01). CONCLUSION Hypoxia modifies the expression of selected SARS-CoV-2 cell entry proteins in human fetal brain endothelial cells in a gestational age- and time-of-exposure-dependent manner. As severe COVID-19 may lead to maternal hypoxia, an altered expression of these proteins in the developing human blood-brain barrier could potentially lead to altered SARS-CoV-2 brain invasion and neurologic sequelae in neonates born to pregnancies complicated by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hafsah Mughis
- Department of Physiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada (Mses Mughis and Lye and Dr Matthews)
| | - Phetcharawan Lye
- Department of Physiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada (Mses Mughis and Lye and Dr Matthews)
| | - Stephen G Matthews
- Department of Physiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada (Mses Mughis and Lye and Dr Matthews); Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (Dr Matthews)
| | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil (Dr Bloise).
| |
Collapse
|
7
|
Loss of Blood-Brain Barrier Integrity in an In Vitro Model Subjected to Intermittent Hypoxia: Is Reversion Possible with a HIF-1α Pathway Inhibitor? Int J Mol Sci 2023; 24:ijms24055062. [PMID: 36902491 PMCID: PMC10003655 DOI: 10.3390/ijms24055062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/11/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Several sleep-related breathing disorders provoke repeated hypoxia stresses, which potentially lead to neurological diseases, such as cognitive impairment. Nevertheless, consequences of repeated intermittent hypoxia on the blood-brain barrier (BBB) are less recognized. This study compared two methods of intermittent hypoxia induction on the cerebral endothelium of the BBB: one using hydralazine and the other using a hypoxia chamber. These cycles were performed on an endothelial cell and astrocyte coculture model. Na-Fl permeability, tight junction protein, and ABC transporters (P-gp and MRP-1) content were evaluated with or without HIF-1 inhibitors YC-1. Our results demonstrated that hydralazine as well as intermittent physical hypoxia progressively altered BBB integrity, as shown by an increase in Na-Fl permeability. This alteration was accompanied by a decrease in concentration of tight junction proteins ZO-1 and claudin-5. In turn, microvascular endothelial cells up-regulated the expression of P-gp and MRP-1. An alteration was also found under hydralazine after the third cycle. On the other hand, the third intermittent hypoxia exposure showed a preservation of BBB characteristics. Furthermore, inhibition of HIF-1α with YC-1 prevented BBB dysfunction after hydralazine treatment. In the case of physical intermittent hypoxia, we observed an incomplete reversion suggesting that other biological mechanisms may be involved in BBB dysfunction. In conclusion, intermittent hypoxia led to an alteration of the BBB model with an adaptation observed after the third cycle.
Collapse
|
8
|
Mani S, Dubey R, Lai IC, Babu MA, Tyagi S, Swargiary G, Mody D, Singh M, Agarwal S, Iqbal D, Kumar S, Hamed M, Sachdeva P, Almutary AG, Albadrani HM, Ojha S, Singh SK, Jha NK. Oxidative Stress and Natural Antioxidants: Back and Forth in the Neurological Mechanisms of Alzheimer's Disease. J Alzheimers Dis 2023; 96:877-912. [PMID: 37927255 DOI: 10.3233/jad-220700] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Alzheimer's disease (AD) is characterized by the progressive degeneration of neuronal cells. With the increase in aged population, there is a prevalence of irreversible neurodegenerative changes, causing a significant mental, social, and economic burden globally. The factors contributing to AD are multidimensional, highly complex, and not completely understood. However, it is widely known that aging, neuroinflammation, and excessive production of reactive oxygen species (ROS), along with other free radicals, substantially contribute to oxidative stress and cell death, which are inextricably linked. While oxidative stress is undeniably important in AD, limiting free radicals and ROS levels is an intriguing and potential strategy for deferring the process of neurodegeneration and alleviating associated symptoms. Therapeutic compounds from natural sources have recently become increasingly accepted and have been effectively studied for AD treatment. These phytocompounds are widely available and a multitude of holistic therapeutic efficiencies for treating AD owing to their antioxidant, anti-inflammatory, and biological activities. Some of these compounds also function by stimulating cholinergic neurotransmission, facilitating the suppression of beta-site amyloid precursor protein-cleaving enzyme 1, α-synuclein, and monoamine oxidase proteins, and deterring the occurrence of AD. Additionally, various phenolic, flavonoid, and terpenoid phytocompounds have been extensively described as potential palliative agents for AD progression. Preclinical studies have shown their involvement in modulating the cellular redox balance and minimizing ROS formation, displaying them as antioxidant agents with neuroprotective abilities. This review emphasizes the mechanistic role of natural products in the treatment of AD and discusses the various pathological hypotheses proposed for AD.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Rajni Dubey
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - I-Chun Lai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Sakshi Tyagi
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Deepansh Mody
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Manisha Singh
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Shriya Agarwal
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Sanjay Kumar
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Munerah Hamed
- Department of Pathology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Hind Muteb Albadrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Kingdom of Saudi Arabia
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | | | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, Uttarakhand, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| |
Collapse
|
9
|
Xue Y, Wang X, Wan B, Wang D, Li M, Cheng K, Luo Q, Wang D, Lu Y, Zhu L. Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema. Cell Commun Signal 2022; 20:160. [PMID: 36253854 PMCID: PMC9575296 DOI: 10.1186/s12964-022-00976-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/18/2022] [Indexed: 11/29/2022] Open
Abstract
Background High-altitude cerebral edema (HACE) is a serious and potentially fatal brain injury that is caused by acute hypobaric hypoxia (HH) exposure. Vasogenic edema is the main pathological factor of this condition. Hypoxia-induced disruptions of tight junctions in the endothelium trigger blood‒brain barrier (BBB) damage and induce vasogenic edema. Nuclear respiratory factor 1 (NRF1) acts as a major regulator of hypoxia-induced endothelial cell injury, and caveolin-1 (CAV-1) is upregulated as its downstream gene in hypoxic endothelial cells. This study aimed to investigate whether CAV-1 is involved in HACE progression and the underlying mechanism. Methods C57BL/6 mice were exposed to HH (7600 m above sea level) for 24 h, and BBB injury was assessed by brain water content, Evans blue staining and FITC-dextran leakage. Immunofluorescence, transmission electron microscope, transendothelial electrical resistance (TEER), transcytosis assays, and western blotting were performed to confirm the role and underlying mechanism of CAV-1 in the disruption of tight junctions and BBB permeability. Mice or bEnd.3 cells were pretreated with MβCD, a specific blocker of CAV-1, and the effect of CAV-1 on claudin-5 internalization under hypoxic conditions was detected by immunofluorescence, western blotting, and TEER. The expression of NRF1 was knocked down, and the regulation of CAV-1 by NRF1 under hypoxic conditions was examined by qPCR, western blotting, and immunofluorescence. Results The BBB was severely damaged and was accompanied by a significant loss of vascular tight junction proteins in HACE mice. CAV-1 was significantly upregulated in endothelial cells, and claudin-5 explicitly colocalized with CAV-1. During the in vitro experiments, hypoxia increased cell permeability, CAV-1 expression, and claudin-5 internalization and downregulated tight junction proteins. Simultaneously, hypoxia induced the upregulation of CAV-1 by activating NRF1. Blocking CAV-1-mediated intracellular transport improved the integrity of TJs in hypoxic endothelial cells and effectively inhibited the increase in BBB permeability and brain water content in HH animals. Conclusions Hypoxia upregulated CAV-1 transcription via the activation of NRF1 in endothelial cells, thus inducing the internalization and autophagic degradation of claudin-5. These effects lead to the destruction of the BBB and trigger HACE. Therefore, CAV-1 may be a potential therapeutic target for HACE. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00976-3.
Collapse
Affiliation(s)
- Yan Xue
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.,Medical School of Nantong University, Nantong, 226007, China.,Nantong Health College of Jiangsu Province, Nantong, 226010, China
| | - Xueting Wang
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Baolan Wan
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Dongzhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Meiqi Li
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Kang Cheng
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Qianqian Luo
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Dan Wang
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Yapeng Lu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.
| |
Collapse
|
10
|
Krakovski MA, Arora N, Jain S, Glover J, Dombrowski K, Hernandez B, Yadav H, Sarma AK. Diet-microbiome-gut-brain nexus in acute and chronic brain injury. Front Neurosci 2022; 16:1002266. [PMID: 36188471 PMCID: PMC9523267 DOI: 10.3389/fnins.2022.1002266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, appreciation for the gut microbiome and its relationship to human health has emerged as a facilitator of maintaining healthy physiology and a contributor to numerous human diseases. The contribution of the microbiome in modulating the gut-brain axis has gained significant attention in recent years, extensively studied in chronic brain injuries such as Epilepsy and Alzheimer’s Disease. Furthermore, there is growing evidence that gut microbiome also contributes to acute brain injuries like stroke(s) and traumatic brain injury. Microbiome-gut-brain communications are bidirectional and involve metabolite production and modulation of immune and neuronal functions. The microbiome plays two distinct roles: it beneficially modulates immune system and neuronal functions; however, abnormalities in the host’s microbiome also exacerbates neuronal damage or delays the recovery from acute injuries. After brain injury, several inflammatory changes, such as the necrosis and apoptosis of neuronal tissue, propagates downward inflammatory signals to disrupt the microbiome homeostasis; however, microbiome dysbiosis impacts the upward signaling to the brain and interferes with recovery in neuronal functions and brain health. Diet is a superlative modulator of microbiome and is known to impact the gut-brain axis, including its influence on acute and neuronal injuries. In this review, we discussed the differential microbiome changes in both acute and chronic brain injuries, as well as the therapeutic importance of modulation by diets and probiotics. We emphasize the mechanistic studies based on animal models and their translational or clinical relationship by reviewing human studies.
Collapse
Affiliation(s)
| | - Niraj Arora
- Department of Neurology, University of Missouri, Columbia, MO, United States
| | - Shalini Jain
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Jennifer Glover
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Keith Dombrowski
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Beverly Hernandez
- Clinical Nutrition Services, Tampa General Hospital, Tampa, FL, United States
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, United States
- *Correspondence: Hariom Yadav,
| | - Anand Karthik Sarma
- Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Neurology, Atrium Health Wake Forest Baptist, Winston-Salem, NC, United States
- Anand Karthik Sarma,
| |
Collapse
|
11
|
Zeng M, Zhang R, Yang Q, Guo L, Zhang X, Yu B, Gan J, Yang Z, Li H, Wang Y, Jiang X, Lu B. Pharmacological therapy to cerebral ischemia-reperfusion injury: Focus on saponins. Biomed Pharmacother 2022; 155:113696. [PMID: 36116247 DOI: 10.1016/j.biopha.2022.113696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Secondary insult from cerebral ischemia-reperfusion injury (CIRI) is a major risk factor for poor prognosis of cerebral ischemia. Saponins are steroid or triterpenoid glycosides with various pharmacological activities that are effective in treating CIRI. By browsing the literature from 2001 to 2021, 55 references involving 24 kinds of saponins were included. Saponins were shown to relieve CIRI by inhibiting oxidation stress, neuroinflammation, and apoptosis, restoring BBB integrity, and promoting neurogenesis and angiogenesis. This review summarizes and classifies several common saponins and their mechanisms in relieving CIRI. Information provided in this review will benefit researchers to design, research and develop new medicines to treat CIRI-related conditions with saponins.
Collapse
Affiliation(s)
- Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiuyue Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bin Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhen Yang
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Bin Lu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
12
|
Kuo YC, Yang IS, Rajesh R. Suppressed XIAP and cIAP expressions in human brain cancer stem cells using BV6- and GDC0152-encapsulated nanoparticles. J Taiwan Inst Chem Eng 2022. [DOI: 10.1016/j.jtice.2022.104394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
13
|
Abstract
SignificancePhylogenies are the basis of many ecological and evolutionary studies. However, zokor phylogeny and speciation patterns are heavily debated. This study disentangled the phylogeny and speciation of zokors genomically. Six species of the Eospalax were separated into high-altitude E. baileyi and E. smithi and the rest four low-altitude species by recent Qinghai-Tibet Plateau uplift 3.6 million y ago. E. rothschildi and E. smithi speciated south of the Qinling-Huaihe Line, where refuges were supplied during glaciation. Introgression and incomplete lineage sorting led to the complex phylogeny of zokors. Genomic islands were formed due to ancient polymorphisms and divergence hitchhiking. This study concluded that climatic, geological, and tectonic events shaped the phylogeny and speciation of zokors in China.
Collapse
|
14
|
Kim Y, Cho AY, Kim HC, Ryu D, Jo SA, Jung YS. Effects of Natural Polyphenols on Oxidative Stress-Mediated Blood–Brain Barrier Dysfunction. Antioxidants (Basel) 2022; 11:antiox11020197. [PMID: 35204080 PMCID: PMC8868362 DOI: 10.3390/antiox11020197] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
The blood-brain barrier (BBB), which consists mainly of brain microvascular endothelial cells and astrocytes connected by tight junctions (TJs) and adhesion molecules (AMs), maintains the homeostatic balance between brain parenchyma and extracellular fluid. Accumulating evidence shows that BBB dysfunction is a common feature of neurodegenerative diseases, including stroke, traumatic brain injury, and Alzheimer’s disease. Among the various pathological pathways of BBB dysfunction, reactive oxygen species (ROS) are known to play a key role in inducing BBB disruption mediated via TJ modification, AM induction, cytoskeletal reorganization, and matrix metalloproteinase activation. Thus, antioxidants have been suggested to exert beneficial effects on BBB dysfunction-associated brain diseases. In this review, we summarized the sources of ROS production in multiple cells that constitute or surround the BBB, such as BBB endothelial cells, astrocytes, microglia, and neutrophils. We also reviewed various pathological mechanisms by which BBB disruption is caused by ROS in these cells. Finally, we summarized the effects of various natural polyphenols on BBB dysfunction to suggest a therapeutic strategy for BBB disruption-related brain diseases.
Collapse
Affiliation(s)
- Yeonjae Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - A Yeon Cho
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
| | - Hong Cheol Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
| | - Dajung Ryu
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - Sangmee Ahn Jo
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea;
- Department of Pharmacology, College of Pharmacy, Dankook University, Cheonan 31116, Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-31-219-3444
| |
Collapse
|
15
|
Akbari Namvar Z, Mahdavi R, Shirmohammadi M, Nikniaz Z. The effect of group-based education on gastrointestinal symptoms and quality of life in patients with celiac disease: randomized controlled clinical trial. BMC Gastroenterol 2022; 22:18. [PMID: 35016615 PMCID: PMC8751319 DOI: 10.1186/s12876-022-02096-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In this trial, we investigated the effect of a group-based education program on gastrointestinal (GI) symptoms and quality of life (QOL) in patients with celiac disease (CD). METHOD In the present study, 130 patients with CD who were on a GFD for at least 3 months, randomly assigned to receive group-based education (n = 66) or routine education in the celiac clinic (n = 64) for 3 months. We assessed gastrointestinal symptoms and quality of life using the gastrointestinal symptom rating scale (GSRS) questionnaire and SF-36 questionnaire at baseline and 3 months after interventions. RESULTS The mean age of the participants was 37.57 ± 9.59 years. There were no significant differences between the two groups regarding the baseline values. Results showed that the mean score of total GSRS score in the intervention group was significantly lower compared with the control group 3 months post-intervention (p = 0.04). Also, there was a significant difference in the mean score of SF-36 between the two groups 3 months post-intervention (p = 0.02). CONCLUSION Results showed that group-based education was an effective intervention in patients with celiac disease to improve gastrointestinal symptoms and quality of life. Trial registration IRCT code: IRCT20080904001197N21; registration date: 5/23/2019.
Collapse
Affiliation(s)
| | - Reza Mahdavi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masood Shirmohammadi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Nikniaz
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Li W, Cao F, Takase H, Arai K, Lo EH, Lok J. Blood-Brain Barrier Mechanisms in Stroke and Trauma. Handb Exp Pharmacol 2022; 273:267-293. [PMID: 33580391 DOI: 10.1007/164_2020_426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The brain microenvironment is tightly regulated. The blood-brain barrier (BBB), which is composed of cerebral endothelial cells, astrocytes, and pericytes, plays an important role in maintaining the brain homeostasis by regulating the transport of both beneficial and detrimental substances between circulating blood and brain parenchyma. After brain injury and disease, BBB tightness becomes dysregulated, thus leading to inflammation and secondary brain damage. In this chapter, we overview the fundamental mechanisms of BBB damage and repair after stroke and traumatic brain injury (TBI). Understanding these mechanisms may lead to therapeutic opportunities for brain injury.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang Cao
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Josephine Lok
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Gong S, Ma H, Zheng F, Huang J, Zhang Y, Yu B, Li F, Kou J. Inhibiting YAP in Endothelial Cells From Entering the Nucleus Attenuates Blood-Brain Barrier Damage During Ischemia-Reperfusion Injury. Front Pharmacol 2021; 12:777680. [PMID: 34899341 PMCID: PMC8662521 DOI: 10.3389/fphar.2021.777680] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Blood-brain barrier (BBB) damage is a critical event in ischemic stroke, contributing to aggravated brain damage. Endothelial cell form a major component of the BBB, but its regulation in stroke has yet to be clarified. We investigated the function of Yes-associated protein 1 (YAP) in the endothelium on BBB breakdown during cerebral ischemia/reperfusion (I/R) injury. The effects of YAP on BBB dysfunction were explored in middle cerebral artery occlusion/reperfusion (MCAO/R)-injury model mice and using brain microvascular endothelial cells (BMEC) exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury. The degree of brain injury was estimated using staining (2,3,5-Triphenyltetrazolium chloride, hematoxylin and eosin) and the detection of cerebral blood flow. BBB breakdown was investigated by examining the leakage of Evans Blue dye and evaluating the expression of tight junction (TJ)-associated proteins and matrix metallopeptidase (MMP) 2 and 9. YAP expression was up-regulated in the nucleus of BMEC after cerebral I/R injury. Verteporfin (YAP inhibitor) down-regulated YAP expression in the nucleus and improved BBB hyperpermeability and TJ integrity disruption stimulated by cerebral I/R. YAP-targeted small interfering RNA (siRNA) exerted the same effects in BMEC cells exposed to OGD/R injury. Our findings provide new insights into the contributions made by YAP to the maintenance of BBB integrity and highlight the potential for YAP to serve as a therapeutic target to modulate BBB integrity following ischemic stroke and related cerebrovascular diseases.
Collapse
Affiliation(s)
- Shuaishuai Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huifen Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fan Zheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Juan Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Boyang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fang Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Junping Kou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Wu L, Islam MR, Lee J, Takase H, Guo S, Andrews AM, Buzhdygan TP, Mathew J, Li W, Arai K, Lo EH, Ramirez SH, Lok J. ErbB3 is a critical regulator of cytoskeletal dynamics in brain microvascular endothelial cells: Implications for vascular remodeling and blood brain barrier modulation. J Cereb Blood Flow Metab 2021; 41:2242-2255. [PMID: 33583260 PMCID: PMC8393293 DOI: 10.1177/0271678x20984976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neuregulin (NRG)1 - ErbB receptor signaling has been shown to play an important role in the biological function of peripheral microvascular endothelial cells. However, little is known about how NRG1/ErbB signaling impacts brain endothelial function and blood-brain barrier (BBB) properties. NRG1/ErbB pathways are affected by brain injury; when brain trauma was induced in mice in a controlled cortical impact model, endothelial ErbB3 gene expression was reduced to a greater extent than that of other NRG1 receptors. This finding suggests that ErbB3-mediated processes may be significantly compromised after injury, and that an understanding of ErbB3 function would be important in the of study of endothelial biology in the healthy and injured brain. Towards this goal, cultured brain microvascular endothelial cells were transfected with siRNA to ErbB3, resulting in alterations in F-actin organization and microtubule assembly, cell morphology, migration and angiogenic processes. Importantly, a significant increase in barrier permeability was observed when ErbB3 was downregulated, suggesting ErbB3 involvement in BBB regulation. Overall, these results indicate that neuregulin-1/ErbB3 signaling is intricately connected with the cytoskeletal processes of the brain endothelium and contributes to morphological and angiogenic changes as well as to BBB integrity.
Collapse
Affiliation(s)
- Limin Wu
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Mohammad R Islam
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Janice Lee
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Hajime Takase
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Shuzhen Guo
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Allison M Andrews
- Department of Pathology & Laboratory Medicine, Temple University School of Medicine, Philadelphia, USA
| | - Tetyana P Buzhdygan
- Department of Pathology & Laboratory Medicine, Temple University School of Medicine, Philadelphia, USA
| | - Justin Mathew
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Wenlu Li
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA.,Department of Radiology, Massachusetts General Hospital, Boston, USA.,Department of Neurology, Massachusetts General Hospital, Boston, USA
| | - Servio H Ramirez
- Department of Pathology & Laboratory Medicine, Temple University School of Medicine, Philadelphia, USA.,The Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA.,Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
19
|
Heidarzadeh M, Gürsoy-Özdemir Y, Kaya M, Eslami Abriz A, Zarebkohan A, Rahbarghazi R, Sokullu E. Exosomal delivery of therapeutic modulators through the blood-brain barrier; promise and pitfalls. Cell Biosci 2021; 11:142. [PMID: 34294165 PMCID: PMC8296716 DOI: 10.1186/s13578-021-00650-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Nowadays, a large population around the world, especially the elderly, suffers from neurological inflammatory and degenerative disorders/diseases. Current drug delivery strategies are facing different challenges because of the presence of the BBB, which limits the transport of various substances and cells to brain parenchyma. Additionally, the low rate of successful cell transplantation to the brain injury sites leads to efforts to find alternative therapies. Stem cell byproducts such as exosomes are touted as natural nano-drug carriers with 50-100 nm in diameter. These nano-sized particles could harbor and transfer a plethora of therapeutic agents and biological cargos to the brain. These nanoparticles would offer a solution to maintain paracrine cell-to-cell communications under healthy and inflammatory conditions. The main question is that the existence of the intact BBB could limit exosomal trafficking. Does BBB possess some molecular mechanisms that facilitate the exosomal delivery compared to the circulating cell? Although preliminary studies have shown that exosomes could cross the BBB, the exact molecular mechanism(s) beyond this phenomenon remains unclear. In this review, we tried to compile some facts about exosome delivery through the BBB and propose some mechanisms that regulate exosomal cross in pathological and physiological conditions.
Collapse
Affiliation(s)
- Morteza Heidarzadeh
- Koç University Research Center for Translational Medicine (KUTTAM), Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Koç University Research Center for Translational Medicine (KUTTAM), Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey.,Neurology Department, Koç University School of Medicine, Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey
| | - Mehmet Kaya
- Koç University Research Center for Translational Medicine (KUTTAM), Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey.,Physiology Department, Koç University School of Medicine, Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey
| | - Aysan Eslami Abriz
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Emel Sokullu
- Koç University Research Center for Translational Medicine (KUTTAM), Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey. .,Biophysics Department, Koç University School of Medicine, Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey.
| |
Collapse
|
20
|
Li X, Tan X, Chen Q, Zhu X, Zhang J, Zhang J, Jia B. Prodigiosin of Serratia marcescens ZPG19 Alters the Gut Microbiota Composition of Kunming Mice. Molecules 2021; 26:molecules26082156. [PMID: 33918541 PMCID: PMC8069934 DOI: 10.3390/molecules26082156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/29/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
Prodigiosin is a red pigment produced by Serratia marcescens with anticancer, antimalarial, and antibacterial effects. In this study, we extracted and identified a red pigment from a culture of S. marcescens strain ZPG19 and investigated its effect on the growth performance and intestinal microbiota of Kunming mice. High-performance liquid chromatography/mass spectrometry revealed that the pigment had a mass-to-charge ratio (m/z) of 324.2160, and thus it was identified as prodigiosin. To investigate the effect of prodigiosin on the intestinal microbiota, mice (n = 5) were administered 150 μg/kg/d prodigiosin (crude extract, 95% purity) via the drinking water for 18 days. Administration of prodigiosin did not cause toxicity in mice. High-throughput sequencing analysis revealed that prodigiosin altered the cecum microbiota abundance and diversity; the relative abundance of Desulfovibrio significantly decreased, whereas Lactobacillus reuteri significantly increased. This finding indicates that oral administration of prodigiosin has a beneficial effect on the intestinal microbiota of mice. As prodigiosin is non-toxic to mouse internal organs and improves the mouse intestinal microbiota, we suggest that it is a promising candidate drug to treat intestinal inflammation.
Collapse
Affiliation(s)
- Xue Li
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250000, China; (X.L.); (X.T.); (Q.C.)
| | - Xinfeng Tan
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250000, China; (X.L.); (X.T.); (Q.C.)
| | - Qingshuang Chen
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250000, China; (X.L.); (X.T.); (Q.C.)
| | - Xiaoling Zhu
- Shandong Academy of Agricultural Sciences, Jinan 250000, China;
| | - Jing Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250000, China; (X.L.); (X.T.); (Q.C.)
- Correspondence: (J.Z.); (J.Z.); (B.J.)
| | - Jie Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250000, China; (X.L.); (X.T.); (Q.C.)
- Correspondence: (J.Z.); (J.Z.); (B.J.)
| | - Baolei Jia
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250000, China; (X.L.); (X.T.); (Q.C.)
- Correspondence: (J.Z.); (J.Z.); (B.J.)
| |
Collapse
|
21
|
Gaur P, Prasad S, Kumar B, Sharma SK, Vats P. High-altitude hypoxia induced reactive oxygen species generation, signaling, and mitigation approaches. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2021; 65:601-615. [PMID: 33156424 DOI: 10.1007/s00484-020-02037-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 10/05/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
Homeostasis between pro-oxidants and anti-oxidants is necessary for aerobic life, which if perturbed and shifted towards pro-oxidants results in oxidative stress. It is generally agreed that reactive oxygen species (ROS) production is accelerated with mountainous elevation, which may play a role in spawning serious health crisis. Exposure to increasing terrestrial altitude leads to a reduction in ambient O2 availability in cells producing a series of hypoxic oxidative stress reactions and altering the redox balance in humans. Enormous literature on redox signaling drove research activity towards understanding the role of oxidative stress under normal and challenging conditions like high-altitude hypoxia which grounds for disturbed redox signaling. Excessive ROS production and accumulation of free radicals in cells and tissues can cause various pulmonary, cardiovascular, and metabolic pathophysiological conditions. In order to counteract this oxidative stress and maintain the balance of pro-oxidants and anti-oxidants, an anti-oxidant system exists in the human body, which, however, gets surpassed by elevated ROS levels, but can be strengthened through the use of anti-oxidant supplements. Such cumulative studies of fundamentals on a global concept like oxidative stress and role of anti-oxidants can act as a foundation to further smoothen for researchers to study over health, disease, and other pathophysiological conditions. This review highlights the interconnection between high altitude and oxidative stress and the role of anti-oxidants to protect cells from oxidative damages and to lower the risk of altitude-associated sickness.
Collapse
Affiliation(s)
- Priya Gaur
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Lucknow Road, Timarpur, Delhi, 110054,, India
| | - Suchita Prasad
- Department of Chemistry, University of Delhi, Delhi, 110007,, India
| | - Bhuvnesh Kumar
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Lucknow Road, Timarpur, Delhi, 110054,, India
| | - Sunil K Sharma
- Department of Chemistry, University of Delhi, Delhi, 110007,, India.
| | - Praveen Vats
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Lucknow Road, Timarpur, Delhi, 110054,, India.
| |
Collapse
|
22
|
Betterton RD, Davis TP, Ronaldson PT. Organic Cation Transporter (OCT/OCTN) Expression at Brain Barrier Sites: Focus on CNS Drug Delivery. Handb Exp Pharmacol 2021; 266:301-328. [PMID: 33674914 PMCID: PMC8603467 DOI: 10.1007/164_2021_448] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Therapeutic delivery to the central nervous system (CNS) continues to be a considerable challenge in the pharmacological treatment and management of neurological disorders. This is primarily due to the physiological and biochemical characteristics of brain barrier sites (i.e., blood-brain barrier (BBB), blood-cerebrospinal fluid barrier (BCSFB)). Drug uptake into brain tissue is highly restricted by expression of tight junction protein complexes and adherens junctions between brain microvascular endothelial cells and choroid plexus epithelial cells. Additionally, efflux transport proteins expressed at the plasma membrane of these same endothelial and epithelial cells act to limit CNS concentrations of centrally acting drugs. In contrast, facilitated diffusion via transporter proteins allows for substrate-specific flux of molecules across the plasma membrane, directing drug uptake into the CNS. Organic Cation Transporters (OCTs) and Novel Organic Cation Transporters (OCTNs) are two subfamilies of the solute carrier 22 (SLC22) family of proteins that have significant potential to mediate delivery of positively charged, zwitterionic, and uncharged therapeutics. While expression of these transporters has been well characterized in peripheral tissues, the functional expression of OCT and OCTN transporters at CNS barrier sites and their role in delivery of therapeutic drugs to molecular targets in the brain require more detailed analysis. In this chapter, we will review current knowledge on localization, function, and regulation of OCT and OCTN isoforms at the BBB and BCSFB with a particular emphasis on how these transporters can be utilized for CNS delivery of therapeutic agents.
Collapse
Affiliation(s)
- Robert D Betterton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
23
|
Ye LX, An NC, Huang P, Li DH, Zheng ZL, Ji H, Li H, Chen DQ, Wu YQ, Xiao J, Xu K, Li XK, Zhang HY. Exogenous platelet-derived growth factor improves neurovascular unit recovery after spinal cord injury. Neural Regen Res 2021; 16:765-771. [PMID: 33063740 PMCID: PMC8067950 DOI: 10.4103/1673-5374.295347] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The blood-spinal cord barrier plays a vital role in recovery after spinal cord injury. The neurovascular unit concept emphasizes the relationship between nerves and vessels in the brain, while the effect of the blood-spinal cord barrier on the neurovascular unit is rarely reported in spinal cord injury studies. Mouse models of spinal cord injury were established by heavy object impact and then immediately injected with platelet-derived growth factor (80 μg/kg) at the injury site. Our results showed that after platelet-derived growth factor administration, spinal cord injury, neuronal apoptosis, and blood-spinal cord barrier permeability were reduced, excessive astrocyte proliferation and the autophagy-related apoptosis signaling pathway were inhibited, collagen synthesis was increased, and mouse locomotor function was improved. In vitro, human umbilical vein endothelial cells were established by exposure to 200 μM H2O2. At 2 hours prior to injury, in vitro cell models were treated with 5 ng/mL platelet-derived growth factor. Our results showed that expression of blood-spinal cord barrier-related proteins, including Occludin, Claudin 5, and β-catenin, was significantly decreased and autophagy was significantly reduced. Additionally, the protective effects of platelet-derived growth factor could be reversed by intraperitoneal injection of 80 mg/kg chloroquine, an autophagy inhibitor, for 3 successive days prior to spinal cord injury. Our findings suggest that platelet-derived growth factor can promote endothelial cell repair by regulating autophagy, improve the function of the blood-spinal cord barrier, and promote the recovery of locomotor function post-spinal cord injury. Approval for animal experiments was obtained from the Animal Ethics Committee, Wenzhou Medical University, China (approval No. wydw2018-0043) in July 2018.
Collapse
Affiliation(s)
- Lu-Xia Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ning-Chen An
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Peng Huang
- Department of Pharmacy, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Duo-Hui Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhi-Long Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hao Ji
- Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang Province, China
| | - Hao Li
- Department of Orthopedics Surgery, Lishui People's Hospital, The sixth affiliated hospital of Wenzhou Medical University, Lishui, Zhejiang Province, China
| | - Da-Qing Chen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yan-Qing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang Province, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ke Xu
- Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang Province, China
| | - Xiao-Kun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hong-Yu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
24
|
Lochhead JJ, Yang J, Ronaldson PT, Davis TP. Structure, Function, and Regulation of the Blood-Brain Barrier Tight Junction in Central Nervous System Disorders. Front Physiol 2020; 11:914. [PMID: 32848858 PMCID: PMC7424030 DOI: 10.3389/fphys.2020.00914] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
The blood-brain barrier (BBB) allows the brain to selectively import nutrients and energy critical to neuronal function while simultaneously excluding neurotoxic substances from the peripheral circulation. In contrast to the highly permeable vasculature present in most organs that reside outside of the central nervous system (CNS), the BBB exhibits a high transendothelial electrical resistance (TEER) along with a low rate of transcytosis and greatly restricted paracellular permeability. The property of low paracellular permeability is controlled by tight junction (TJ) protein complexes that seal the paracellular route between apposing brain microvascular endothelial cells. Although tight junction protein complexes are principal contributors to physical barrier properties, they are not static in nature. Rather, tight junction protein complexes are highly dynamic structures, where expression and/or localization of individual constituent proteins can be modified in response to pathophysiological stressors. These stressors induce modifications to tight junction protein complexes that involve de novo synthesis of new protein or discrete trafficking mechanisms. Such responsiveness of BBB tight junctions to diseases indicates that these protein complexes are critical for maintenance of CNS homeostasis. In fulfillment of this vital role, BBB tight junctions are also a major obstacle to therapeutic drug delivery to the brain. There is an opportunity to overcome this substantial obstacle and optimize neuropharmacology via acquisition of a detailed understanding of BBB tight junction structure, function, and regulation. In this review, we discuss physiological characteristics of tight junction protein complexes and how these properties regulate delivery of therapeutics to the CNS for treatment of neurological diseases. Specifically, we will discuss modulation of tight junction structure, function, and regulation both in the context of disease states and in the setting of pharmacotherapy. In particular, we will highlight how these properties can be potentially manipulated at the molecular level to increase CNS drug levels via paracellular transport to the brain.
Collapse
|
25
|
Oxidative Stress-Mediated Blood-Brain Barrier (BBB) Disruption in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020. [DOI: 10.1155/2020/4356386] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB), as a crucial gate of brain-blood molecular exchange, is involved in the pathogenesis of multiple neurological diseases. Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) and the scavenger system. Since oxidative stress plays a significant role in the production and maintenance of the BBB, the cerebrovascular system is especially vulnerable to it. The pathways that initiate BBB dysfunction include, but are not limited to, mitochondrial dysfunction, excitotoxicity, iron metabolism, cytokines, pyroptosis, and necroptosis, all converging on the generation of ROS. Interestingly, ROS also provide common triggers that directly regulate BBB damage, parameters including tight junction (TJ) modifications, transporters, matrix metalloproteinase (MMP) activation, inflammatory responses, and autophagy. We will discuss the role of oxidative stress-mediated BBB disruption in neurological diseases, such as hemorrhagic stroke, ischemic stroke (IS), Alzheimer’s disease (AD), Parkinson’s disease (PD), traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), and cerebral small vessel disease (CSVD). This review will also discuss the latest clinical evidence of potential biomarkers and antioxidant drugs towards oxidative stress in neurological diseases. A deeper understanding of how oxidative stress damages BBB may open up more therapeutic options for the treatment of neurological diseases.
Collapse
|
26
|
Liktor-Busa E, Blawn KT, Kellohen KL, Wiese BM, Verkhovsky V, Wahl J, Vivek A, Palomino SM, Davis TP, Vanderah TW, Largent-Milnes TM. Functional NHE1 expression is critical to blood brain barrier integrity and sumatriptan blood to brain uptake. PLoS One 2020; 15:e0227463. [PMID: 32469979 PMCID: PMC7259629 DOI: 10.1371/journal.pone.0227463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/04/2020] [Indexed: 12/31/2022] Open
Abstract
Disruption of blood-brain barrier integrity and dramatic failure of brain ion homeostasis including fluctuations of pH occurs during cortical spreading depression (CSD) events associated with several neurological disorders, including migraine with aura, traumatic brain injury and stroke. NHE1 is the primary regulator of pH in the central nervous system. The goal of the current study was to investigate the role of sodium-hydrogen exchanger type 1 (NHE1) in blood brain barrier (BBB) integrity during CSD events and the contributions of this antiporter on xenobiotic uptake. Using immortalized cell lines, pharmacologic inhibition and genetic knockdown of NHE1 mitigated the paracellular uptake of radiolabeled sucrose implicating functional NHE1 in BBB maintenance. In contrast, loss of functional NHE1 in endothelial cells facilitated uptake of the anti-migraine therapeutic, sumatriptan. In female rats, cortical KCl but not aCSF selectively reduced total expression of NHE1 in cortex and PAG but increased expression in trigeminal ganglia; no changes were seen in trigeminal nucleus caudalis. Thus, in vitro observations may have a significance in vivo to increase brain sumatriptan levels. Pharmacological inhibition of NHE1 prior to cortical manipulations enhanced the efficacy of sumatriptan at early time-points but induced facial sensitivity alone. Overall, our results suggest that dysregulation of NHE1 contributes to breaches in BBB integrity, drug penetrance, and the behavioral sensitivity to the antimigraine agent, sumatriptan.
Collapse
Affiliation(s)
- Erika Liktor-Busa
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Kiera T. Blawn
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Kathryn L. Kellohen
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Beth M. Wiese
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Vani Verkhovsky
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Jared Wahl
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Anjali Vivek
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Seph M. Palomino
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Todd W. Vanderah
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - Tally M. Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
27
|
Robinson BD, Tharakan B, Lomas A, Wiggins-Dohlvik K, Alluri H, Shaji CA, Jupiter D, Isbell CL. Exploring blood-brain barrier hyperpermeability and potential biomarkers in traumatic brain injury. Proc (Bayl Univ Med Cent) 2020; 33:199-204. [PMID: 32313461 DOI: 10.1080/08998280.2020.1727706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 01/01/2023] Open
Abstract
Blood-brain barrier breakdown and associated vascular hyperpermeability leads to vasogenic edema in traumatic brain injury (TBI). Tight junctions maintain blood-brain barrier integrity; their disruption in TBI holds significant promise for diagnosis and treatment. A controlled cortical impactor was used for TBI in mouse studies. Blood was collected 1 h after injury and sent for antibody microarray analysis. Twenty human subjects with radiographic evidence of TBI were enrolled and blood collected within 48 h of admission. Control subjects were individuals with nontrauma diagnoses. The subjects were matched by age and gender. Enzyme-linked immunosorbent assays were performed on each TBI and control sample for tight junction-associated proteins (TJPs), inflammatory markers, and S100β. Plasma was used to conduct in vitro monolayer permeability studies with human brain endothelial cells. S100β and the TJP occludin were significantly elevated in TBI plasma in both the murine and human studies. Monolayer permeability studies showed increased hyperpermeability in TBI groups. Plasma from TBI subjects increases microvascular hyperpermeability in vitro. TJPs in the blood may be a potential biomarker for TBI.
Collapse
Affiliation(s)
| | - Binu Tharakan
- Department of Surgery, Baylor Scott and White Medical CenterTempleTexas.,School of Medicine, Texas A&M Health Sciences CenterTempleTexas
| | - Angela Lomas
- School of Medicine, Texas A&M Health Sciences CenterTempleTexas
| | | | | | | | - Daniel Jupiter
- Department of Preventive Medicine and Community Health, University of Texas Medical BranchGalvestonTexas
| | | |
Collapse
|
28
|
Humer E, Probst T, Pieh C. Metabolomics in Psychiatric Disorders: What We Learn from Animal Models. Metabolites 2020; 10:E72. [PMID: 32079262 PMCID: PMC7074444 DOI: 10.3390/metabo10020072] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Biomarkers are a recent research target within biological factors of psychiatric disorders. There is growing evidence for deriving biomarkers within psychiatric disorders in serum or urine samples in humans, however, few studies have investigated this differentiation in brain or cerebral fluid samples in psychiatric disorders. As brain samples from humans are only available at autopsy, animal models are commonly applied to determine the pathogenesis of psychiatric diseases and to test treatment strategies. The aim of this review is to summarize studies on biomarkers in animal models for psychiatric disorders. For depression, anxiety and addiction disorders studies, biomarkers in animal brains are available. Furthermore, several studies have investigated psychiatric medication, e.g., antipsychotics, antidepressants, or mood stabilizers, in animals. The most notable changes in biomarkers in depressed animal models were related to the glutamate-γ-aminobutyric acid-glutamine-cycle. In anxiety models, alterations in amino acid and energy metabolism (i.e., mitochondrial regulation) were observed. Addicted animals showed several biomarkers according to the induced drugs. In summary, animal models provide some direct insights into the cellular metabolites that are produced during psychiatric processes. In addition, the influence on biomarkers due to short- or long-term medication is a noticeable finding. Further studies should combine representative animal models and human studies on cerebral fluid to improve insight into mental disorders and advance the development of novel treatment strategies.
Collapse
Affiliation(s)
- Elke Humer
- Department for Psychotherapy and Biopsychosocial Health, Danube University Krems, 3500 Krems, Austria; (T.P.); (C.P.)
| | | | | |
Collapse
|
29
|
Williams EI, Betterton RD, Davis TP, Ronaldson PT. Transporter-Mediated Delivery of Small Molecule Drugs to the Brain: A Critical Mechanism That Can Advance Therapeutic Development for Ischemic Stroke. Pharmaceutics 2020; 12:pharmaceutics12020154. [PMID: 32075088 PMCID: PMC7076465 DOI: 10.3390/pharmaceutics12020154] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemic stroke is the 5th leading cause of death in the United States. Despite significant improvements in reperfusion therapies, stroke patients still suffer from debilitating neurocognitive deficits. This indicates an essential need to develop novel stroke treatment paradigms. Endogenous uptake transporters expressed at the blood-brain barrier (BBB) provide an excellent opportunity to advance stroke therapy via optimization of small molecule neuroprotective drug delivery to the brain. Examples of such uptake transporters include organic anion transporting polypeptides (OATPs in humans; Oatps in rodents) and organic cation transporters (OCTs in humans; Octs in rodents). Of particular note, small molecule drugs that have neuroprotective properties are known substrates for these transporters and include 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (i.e., statins) for OATPs/Oatps and 1-amino-3,5-dimethyladamantane (i.e., memantine) for OCTs/Octs. Here, we review current knowledge on specific BBB transporters that can be targeted for improvement of ischemic stroke treatment and provide state-of-the-art perspectives on the rationale for considering BBB transport properties during discovery/development of stroke therapeutics.
Collapse
|
30
|
Abstract
Knowledge about the transport of active compounds across the blood-brain barrier is of essential importance for drug development. Systemically applied drugs for the central nervous system (CNS) must be able to cross the blood-brain barrier in order to reach their target sites, whereas drugs that are supposed to act in the periphery should not permeate the blood-brain barrier so that they do not trigger any adverse central adverse effects. A number of approaches have been pursued, and manifold in silico, in vitro, and in vivo animal models were developed in order to be able to make a better prediction for humans about the possible penetration of active substances into the CNS. In this particular case, however, in vitro models play a special role, since the data basis for in silico models is usually in need of improvement, and the predictive power of in vivo animal models has to be checked for possible species differences. The blood-brain barrier is a dynamic, highly selective barrier formed by brain capillary endothelial cells. One of its main tasks is the maintenance of homeostasis in the CNS. The function of the barrier is regulated by cells of the microenvironment and the shear stress mediated by the blood flow, which makes the model development most complex. In general, one could follow the credo "as easy as possible, as complex as necessary" for the usage of in vitro BBB models for drug development. In addition to the description of the classical cell culture models (transwell, hollow fiber) and guidance how to apply them, the latest developments (spheroids, microfluidic models) will be introduced in this chapter, as it is attempted to get more in vivo-like and to be applicable for high-throughput usage with these models. Moreover, details about the development of models based on stem cells derived from different sources with a special focus on human induced pluripotent stem cells are presented.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Competence Unit Molecular Diagnostics, Center Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria.
| |
Collapse
|
31
|
DING Y, LI W, WANG R, ZHANG J. [Research progress on the effects of plateau hypoxia on blood-brain barrier structure and drug permeability]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:668-673. [PMID: 31955542 PMCID: PMC8800771 DOI: 10.3785/j.issn.1008-9292.2019.12.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/03/2019] [Indexed: 06/10/2023]
Abstract
Drugs for the treatment of central nervous system diseases need to enter the brain tissue through the blood-brain barrier to function. In high altitude hypoxic environment, there are changes in tight junction proteins of blood-brain barrier tissue structure, transporters in astrocytes and endothelial cells and ATP in endothelial cells; at the same time the permeability of the blood-brain barrier is increased. These changes are an important reference for rational drug use in patients with central nervous system disease in the plateau region. This article reviews the research progress on the effects of plateau hypoxia on the structure of the blood-brain barrier and related drug permeability.
Collapse
Affiliation(s)
| | | | - Rong WANG
- 王荣(1969-), 男, 博士, 主任药师, 教授, 博士生导师, 主要从事高原药代动力学等研究, E-mail:
;
https://orcid.org/0000-0001-9139-7311
| | - Jianchun ZHANG
- 张建春(1968-), 女, 博士, 硕士生导师, 主任药师, 主要从事药物新剂型及中药制剂研究, E-mail:
;
https://orcid.org/0000-0001-9459-8995
| |
Collapse
|
32
|
Bernard-Patrzynski F, Lécuyer MA, Puscas I, Boukhatem I, Charabati M, Bourbonnière L, Ramassamy C, Leclair G, Prat A, Roullin VG. Isolation of endothelial cells, pericytes and astrocytes from mouse brain. PLoS One 2019; 14:e0226302. [PMID: 31851695 PMCID: PMC6919623 DOI: 10.1371/journal.pone.0226302] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/22/2019] [Indexed: 11/24/2022] Open
Abstract
Primary cell isolation from the central nervous system (CNS) has allowed fundamental understanding of blood-brain barrier (BBB) properties. However, poorly described isolation techniques or suboptimal cellular purity has been a weak point of some published scientific articles. Here, we describe in detail how to isolate and enrich, using a common approach, endothelial cells (ECs) from adult mouse brains, as well as pericytes (PCs) and astrocytes (ACs) from newborn mouse brains. Our approach allowed the isolation of these three brain cell types with purities of around 90%. Furthermore, using our protocols, around 3 times more PCs and 2 times more ACs could be grown in culture, as compared to previously published protocols. The cells were identified and characterized using flow cytometry and confocal microscopy. The ability of ECs to form a tight monolayer was assessed for passages 0 to 3. The expression of claudin-5, occludin, zonula occludens-1, P-glycoprotein-1 and breast cancer resistance protein by ECs, as well as the ability of the cells to respond to cytokine stimuli (TNF-α, IFN-γ) was also investigated by q-PCR. The transcellular permeability of ECs was evaluated in the presence of pericytes or astrocytes in a Transwell® model by measuring the transendothelial electrical resistance (TEER), dextran-FITC and sodium fluorescein permeability. Overall, ECs at passages 0 and 1 featured the best properties valued in a BBB model. Furthermore, pericytes did not increase tightness of EC monolayers, whereas astrocytes did regardless of their seeding location. Finally, ECs resuspended in fetal bovine serum (FBS) and dimethyl sulfoxide (DMSO) could be cryopreserved in liquid nitrogen without affecting their phenotype nor their capacity to form a tight monolayer, thus allowing these primary cells to be used for various longitudinal in vitro studies of the blood-brain barrier.
Collapse
Affiliation(s)
| | - Marc-André Lécuyer
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, Göttingen, Germany
| | - Ina Puscas
- Faculty of Pharmacy, Université de Montréal, Montreal, Québec, Canada
| | - Imane Boukhatem
- Faculty of Pharmacy, Université de Montréal, Montreal, Québec, Canada
| | - Marc Charabati
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Lyne Bourbonnière
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Charles Ramassamy
- Institut National de la Recherche Scientifique, Armand-Frappier Institute, Laval, Québec, Canada
| | - Grégoire Leclair
- Faculty of Pharmacy, Université de Montréal, Montreal, Québec, Canada
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - V Gaëlle Roullin
- Faculty of Pharmacy, Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
33
|
Perivascular and Perineural Pathways Involved in Brain Delivery and Distribution of Drugs after Intranasal Administration. Pharmaceutics 2019; 11:pharmaceutics11110598. [PMID: 31726721 PMCID: PMC6921024 DOI: 10.3390/pharmaceutics11110598] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022] Open
Abstract
One of the most challenging aspects of treating disorders of the central nervous system (CNS) is the efficient delivery of drugs to their targets within the brain. Only a small fraction of drugs is able to cross the blood–brain barrier (BBB) under physiological conditions, and this observation has prompted investigation into the routes of administration that may potentially bypass the BBB and deliver drugs directly to the CNS. One such route is the intranasal (IN) route. Increasing evidence has suggested that intranasally-administered drugs are able to bypass the BBB and access the brain through anatomical pathways connecting the nasal cavity to the CNS. Though the exact mechanisms regulating the delivery of therapeutics following IN administration are not fully understood, current evidence suggests that the perineural and perivascular spaces of the olfactory and trigeminal nerves are involved in brain delivery and cerebral perivascular spaces are involved in widespread brain distribution. Here, we review evidence for these delivery and distribution pathways, and we address questions that should be resolved in order to optimize the IN route of administration as a viable strategy to treat CNS disease states.
Collapse
|
34
|
Xia QP, Cheng ZY, He L. The modulatory role of dopamine receptors in brain neuroinflammation. Int Immunopharmacol 2019; 76:105908. [PMID: 31622861 DOI: 10.1016/j.intimp.2019.105908] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 01/11/2023]
Abstract
Neuroinflammation is a general pathological feature of central nervous system (CNS) diseases, primarily caused by activation of astrocytes and microglia, as well as the infiltration of peripheral immune cells. Inhibition of neuroinflammation is an important strategy in the treatment of brain disorders. Dopamine (DA) receptor, a significant G protein-coupled receptor (GPCR), is classified into two families: D1-like (D1 and D5) and D2-like (D2, D3 and D4) receptor families, according to their downstream signaling pathways. Traditionally, DA receptor forms a wide variety of psychological activities and motor functions, such as voluntary movement, working memory and learning. Recently, the role of DA receptor in neuroinflammation has been investigated widely, mainly focusing on nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome, renin-angiotensin system, αB-crystallin, as well as invading peripheral immune cells, including T cells, dendritic cells, macrophages and monocytes. This review briefly outlined the functions and signaling pathways of DA receptor subtypes as well as its role in inflammation-related glial cells, and subsequently summarized the mechanisms of DA receptors affecting neuroinflammation. Meaningfully, this article provided a theoretical basis for drug development targeting DA receptors in inflammation-related brain diseases.
Collapse
Affiliation(s)
- Qing-Peng Xia
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhao-Yan Cheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
35
|
Oxidized Cell-Free DNA Role in the Antioxidant Defense Mechanisms under Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1245749. [PMID: 31360293 PMCID: PMC6644271 DOI: 10.1155/2019/1245749] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/08/2019] [Indexed: 12/15/2022]
Abstract
The present study focuses on the investigation of the oxidized cell-free DNA (cfDNA) properties in several experimental models, including cultured cerebellum cells, peripheral blood lymphocytes (PBL), plasma, and hippocampus under an acute and chronic unpredictable stress model in rats. Firstly, our study shows that Spectrum Green fluorescence-labeled oxidized cfDNA fragments were transferred into the cytoplasm of 80% of the cerebellum culture cells; meanwhile, the nonoxidized cfDNA fragments do not pass into the cells. Oxidized cfDNA stimulates the antioxidant mechanisms and induction of transcription factor NRF2 expression, followed by an activation of NRF2 signaling pathway genes-rise of Nrf2 and Hmox1 gene expression and consequently NRF2 protein synthesis. Secondly, we showed that stress increases plasma cfDNA concentration in rats corresponding with the duration of the stress exposure. At the same time, our study did not reveal any significant changes of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) level in PBL of rats under acute or chronic stress, probably due to the significantly increased Nrf2 expression, that we found in such conditions. 8-oxodG is one of the most reliable markers of DNA oxidation. We also found an increased level of 8-oxodG in the hippocampal homogenates and hippocampal dentate gyrus in rats subjected to acute and chronic stress. Taken together, our data shows that oxidized cfDNA may play a significant role in systemic and neuronal physiological mechanisms of stress and adaptation.
Collapse
|
36
|
Xu H, Liu Y, Wang D, Zhang Z. Shenmai injection maintains blood-brain barrier integrity following focal cerebral ischemia via modulating the expression and trafficking of occludin in lipid rafts. JOURNAL OF ETHNOPHARMACOLOGY 2019; 237:55-63. [PMID: 30902744 DOI: 10.1016/j.jep.2019.03.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/20/2019] [Accepted: 03/11/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenmai injection (SMI), a traditional Chinese herbal medicine is widely used for the clinical treatment of cerebral infarction in China. AIM OF THE STUDY Tight junctions (TJs) are major components of the blood-brain barrier (BBB) that physically restrict the paracellular diffusion of blood-borne substances between endothelial cells into the CNS. TJ proteins are associated with cholesterol-enriched regions of plasma membrane known as lipid rafts, which are critical for the trafficking, positioning and function of TJ proteins. In this study, we investigated the effect of SMI on the expression and trafficking of the key TJ-associated protein, occludin, in lipid rafts. MATERIALS AND METHODS Using a neutral pH, rat cerebral microvessels were subjected to detergent-free density-gradient fractionation to isolate lipid rafts containing occludin. Transmission electron microscopy (TEM) was performed to study the effects of drug administration on ultrastructural changes to TJs. Western blotting (WB), immunofluorescence (IF), and co-immunoprecipitation (COIP) were used to observe the localization and function of TJ-associated proteins. RESULTS We successfully isolated cerebral microvessels and separated lipid rafts from plasma membranes. With SMI treatment, extravasation of FITC-albumin decreased around the cerebral vessels by IF, the tight junctions were found to still be intact and the basement membrane appeared to be of uniform thickness in TEM. Compared with the untreated group, the co-expression of flotillin-1 and occludin in microvascular endothelial cells was increased and distributed continuously in SMI treatment as shown in double label IF. SMI significantly increased the translocation of occludin to lipid raft fractions by WB and COIP. CONCLUSIONS SMI helps maintain the proper assembly of the TJ multiprotein complex in lipid rafts, thereby helping to preserve BBB functional integrity during focal cerebral ischemic insult. Our findings enhance our understanding of the mechanisms underlying the neuroprotective effect of SMI in cerebral ischemia.
Collapse
Affiliation(s)
- Huaming Xu
- Department of Integrated Traditional and Western Medicine, Xiangya Hospital of Central South University, No.87 Xiangya Road, Changsha, Hunan Province, 410008, China; Henan University of Chinese Medicine, No.156 Jinshui East Road, Zhengzhou, Henan Province, 450046, China.
| | - Yuan Liu
- Beijing Hongci Healthcare Investment Management Co., Ltd., 89 Jinbao Street, Dongcheng District, Beijing, 100005, China.
| | - Dongsheng Wang
- Department of Integrated Traditional and Western Medicine, Xiangya Hospital of Central South University, No.87 Xiangya Road, Changsha, Hunan Province, 410008, China.
| | - Zhenqiang Zhang
- Henan University of Chinese Medicine, No.156 Jinshui East Road, Zhengzhou, Henan Province, 450046, China.
| |
Collapse
|
37
|
Cui D, Arima M, Hirayama T, Ikeda E. Hypoxia-induced disruption of neural vascular barrier is mediated by the intracellular induction of Fe(II) ion. Exp Cell Res 2019; 379:166-171. [PMID: 30970238 DOI: 10.1016/j.yexcr.2019.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/29/2022]
Abstract
Neural vascular barrier maintains the optimal tissue microenvironment of central nervous system in which neural cells can function normally. In various neural diseases, the decrease in oxygen concentration, hypoxia, of affected tissues is known to accelerate the disease progression through disruption of neural vascular barrier. Therefore, the clarification of mechanisms underlying hypoxia-induced disruption of neural vascular barrier would definitely lead to the establishment of new effective therapies for intractable neural diseases. In the present study, we first found that hypoxia disrupts neural vascular barrier through pathways independent of HIF-1α and HIF-2α. Then, with a specific fluorescence probe for ferrous, Fe(II) ion, we have obtained the interesting data showing that hypoxia increased the intracellular level of Fe(II) ion in endothelial cells of our in vitro model for neural vascular barrier, and that hypoxia-induced disruption of neural vascular barrier could be inhibited by chelating Fe(II) ion in endothelial cells. Furthermore, in the presence of a reducing reagent for reactive oxygen species (ROS), hypoxia could not disrupt the neural vascular barrier despite that the hypoxic increase in intracellular level of Fe(II) ion was confirmed in endothelial cells. These results indicate that hypoxia-triggered increase in the level of intracellular Fe(II) ion and subsequent production of ROS, probably through Fenton reaction, are the essential pathway mediating the disruption of neural vascular barrier under hypoxia.
Collapse
Affiliation(s)
- Dan Cui
- Department of Pathology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Mitsuru Arima
- Department of Pathology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan; Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4, Daigaku-nishi, Gifu-shi, Gifu, 501-1196, Japan
| | - Eiji Ikeda
- Department of Pathology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
38
|
Kempuraj D, Mentor S, Thangavel R, Ahmed ME, Selvakumar GP, Raikwar SP, Dubova I, Zaheer S, Iyer SS, Zaheer A. Mast Cells in Stress, Pain, Blood-Brain Barrier, Neuroinflammation and Alzheimer's Disease. Front Cell Neurosci 2019; 13:54. [PMID: 30837843 PMCID: PMC6389675 DOI: 10.3389/fncel.2019.00054] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Mast cell activation plays an important role in stress-mediated disease pathogenesis. Chronic stress cause or exacerbate aging and age-dependent neurodegenerative diseases. The severity of inflammatory diseases is worsened by the stress. Mast cell activation-dependent inflammatory mediators augment stress associated pain and neuroinflammation. Stress is the second most common trigger of headache due to mast cell activation. Alzheimer's disease (AD) is a progressive irreversible neurodegenerative disease that affects more women than men and woman's increased susceptibility to chronic stress could increase the risk for AD. Modern life-related stress, social stress, isolation stress, restraint stress, early life stress are associated with an increased level of neurotoxic beta amyloid (Aβ) peptide. Stress increases cognitive dysfunction, generates amyloid precursor protein (APP), hyperphosphorylated tau, neurofibrillary tangles (NFTs), and amyloid plaques (APs) in the brain. Stress-induced Aβ persists for years and generates APs even several years after the stress exposure. Stress activates hypothalamic-pituitary adrenal (HPA) axis and releases corticotropin-releasing hormone (CRH) from hypothalamus and in peripheral system, which increases the formation of Aβ, tau hyperphosphorylation, and blood-brain barrier (BBB) disruption in the brain. Mast cells are implicated in nociception and pain. Mast cells are the source and target of CRH and other neuropeptides that mediate neuroinflammation. Microglia express receptor for CRH that mediate neurodegeneration in AD. However, the exact mechanisms of how stress-mediated mast cell activation contribute to the pathogenesis of AD remains elusive. This mini-review highlights the possible role of stress and mast cell activation in neuroinflammation, BBB, and tight junction disruption and AD pathogenesis.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Shireen Mentor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Mohammad E. Ahmed
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sudhanshu P. Raikwar
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Iuliia Dubova
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Shankar S. Iyer
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans’ Hospital (VA), U.S. Department of Veterans Affairs, Columbia, MO, United States
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
39
|
Anasooya Shaji C, Robinson BD, Yeager A, Beeram MR, Davis ML, Isbell CL, Huang JH, Tharakan B. The Tri-phasic Role of Hydrogen Peroxide in Blood-Brain Barrier Endothelial cells. Sci Rep 2019; 9:133. [PMID: 30644421 PMCID: PMC6333800 DOI: 10.1038/s41598-018-36769-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Hydrogen peroxide (H2O2) plays an important role physiologically as the second messenger and pathologically as an inducer of oxidative stress in injury, ischemia and other conditions. However, it is unclear how H2O2 influences various cellular functions in health and disease differentially, particularly in the blood-brain barrier (BBB). We hypothesized that the change in cellular concentrations of H2O2 is a major contributor in regulation of angiogenesis, barrier integrity/permeability and cell death/apoptosis in BBB endothelial cells. Rat brain microvascular endothelial cells were exposed to various concentrations of H2O2 (1 nM to 25 mM). BBB tight junction protein (zonula ocludens-1; ZO-1) localization and expression, cytoskeletal organization, monolayer permeability, angiogenesis, cell viability and apoptosis were evaluated. H2O2 at low concentrations (0.001 μM to 1 μM) increased endothelial cell tube formation indicating enhanced angiogenesis. H2O2 at 100 μM and above induced monolayer hyperpermeability significantly (p < 0.05). H2O2 at 10 mM and above decreased cell viability and induced apoptosis (p < 0.05). There was a decrease of ZO-1 tight junction localization with 100 μm H2O2, but had no effect on protein expression. Cytoskeletal disorganizations were observed starting at 1 μm. In conclusion H2O2 influences angiogenesis, permeability, and cell death/apoptosis in a tri-phasic and concentration-dependent manner in microvascular endothelial cells of the blood-brain barrier.
Collapse
Affiliation(s)
- Chinchusha Anasooya Shaji
- Department of Surgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Bobby D Robinson
- Department of Surgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Antonia Yeager
- Department of Surgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Madhava R Beeram
- Department of Pediatrics, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Matthew L Davis
- Department of Surgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Claire L Isbell
- Department of Surgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Jason H Huang
- Department of Neurosurgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA
| | - Binu Tharakan
- Department of Surgery, Texas A&M University Health Science Center College of Medicine and Baylor Scott & White Health, Temple, Texas, USA.
| |
Collapse
|
40
|
Morrison HW, Filosa JA. Stroke and the neurovascular unit: glial cells, sex differences, and hypertension. Am J Physiol Cell Physiol 2019; 316:C325-C339. [PMID: 30601672 DOI: 10.1152/ajpcell.00333.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A functional neurovascular unit (NVU) is central to meeting the brain's dynamic metabolic needs. Poststroke damage to the NVU within the ipsilateral hemisphere ranges from cell dysfunction to complete cell loss. Thus, understanding poststroke cell-cell communication within the NVU is of critical importance. Loss of coordinated NVU function exacerbates ischemic injury. However, particular cells of the NVU (e.g., astrocytes) and those with ancillary roles (e.g., microglia) also contribute to repair mechanisms. Epidemiological studies support the notion that infarct size and recovery outcomes are heterogeneous and greatly influenced by modifiable and nonmodifiable factors such as sex and the co-morbid condition common to stroke: hypertension. The mechanisms whereby sex and hypertension modulate NVU function are explored, to some extent, in preclinical laboratory studies. We present a review of the NVU in the context of ischemic stroke with a focus on glial contributions to NVU function and dysfunction. We explore the impact of sex and hypertension as modifiable and nonmodifiable risk factors and the underlying cellular mechanisms that may underlie heterogeneous stroke outcomes. Most of the preclinical investigative studies of poststroke NVU dysfunction are carried out primarily in male stroke models lacking underlying co-morbid conditions, which is very different from the human condition. As such, the evolution of translational medicine to target the NVU for improved stroke outcomes remains elusive; however, it is attainable with further research.
Collapse
|
41
|
Deng FL, Pan JX, Zheng P, Xia JJ, Yin BM, Liang WW, Li YF, Wu J, Xu F, Wu QY, Qu CH, Li W, Wang HY, Xie P. Metabonomics reveals peripheral and central short-chain fatty acid and amino acid dysfunction in a naturally occurring depressive model of macaques. Neuropsychiatr Dis Treat 2019; 15:1077-1088. [PMID: 31118641 PMCID: PMC6501704 DOI: 10.2147/ndt.s186071] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Depression is a complex psychiatric disorder. Various depressive rodent models are usually constructed based on different pathogenesis hypotheses. MATERIALS AND METHODS Herein, using our previously established naturally occurring depressive (NOD) model in a non-human primate (cynomolgus monkey, Macaca fascularis), we performed metabolomics analysis of cerebrospinal fluid (CSF) from NOD female macaques (N=10) and age-and gender-matched healthy controls (HCs) (N=12). Multivariate statistical analysis was used to identify the differentially expressed metabolites between the two groups. Ingenuity Pathways Analysis and MetaboAnalyst were applied for predicted pathways and biological functions analysis. RESULTS Totally, 37 metabolites responsible for discriminating the two groups were identified. The NOD macaques were mainly characterized by perturbations of fatty acid biosynthesis, ABC transport system, and amino acid metabolism (eg, aspartate, glycine, serine, and threonine metabolism). Interestingly, we found that eight altered CSF metabolites belonging to short-chain fatty acids and amino acids were also observed in the serum of NOD macaques (N=13 per group). CONCLUSION Our findings suggest that peripheral and central short-chain fatty acids and amino acids are implicated in the onset of depression.
Collapse
Affiliation(s)
- Feng-Li Deng
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun-Xi Pan
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,The First Affiliated Hospital of Kunming Medical University, Kunming 650032, People's Republic of China
| | - Peng Zheng
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jin-Jun Xia
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Bang-Min Yin
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Wei-Wei Liang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Yi-Fan Li
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jing Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Fan Xu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Qing-Yuan Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China, .,Department of Neurology, Three Gorges Central Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chao-Hua Qu
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Wei Li
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402460, People's Republic of China, .,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China, .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, People's Republic of China,
| |
Collapse
|
42
|
Modulation of Opioid Transport at the Blood-Brain Barrier by Altered ATP-Binding Cassette (ABC) Transporter Expression and Activity. Pharmaceutics 2018; 10:pharmaceutics10040192. [PMID: 30340346 PMCID: PMC6321372 DOI: 10.3390/pharmaceutics10040192] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 12/23/2022] Open
Abstract
Opioids are highly effective analgesics that have a serious potential for adverse drug reactions and for development of addiction and tolerance. Since the use of opioids has escalated in recent years, it is increasingly important to understand biological mechanisms that can increase the probability of opioid-associated adverse events occurring in patient populations. This is emphasized by the current opioid epidemic in the United States where opioid analgesics are frequently abused and misused. It has been established that the effectiveness of opioids is maximized when these drugs readily access opioid receptors in the central nervous system (CNS). Indeed, opioid delivery to the brain is significantly influenced by the blood-brain barrier (BBB). In particular, ATP-binding cassette (ABC) transporters that are endogenously expressed at the BBB are critical determinants of CNS opioid penetration. In this review, we will discuss current knowledge on the transport of opioid analgesic drugs by ABC transporters at the BBB. We will also examine how expression and trafficking of ABC transporters can be modified by pain and/or opioid pharmacotherapy, a novel mechanism that can promote opioid-associated adverse drug events and development of addiction and tolerance.
Collapse
|
43
|
Obrenovich MEM. Leaky Gut, Leaky Brain? Microorganisms 2018; 6:microorganisms6040107. [PMID: 30340384 PMCID: PMC6313445 DOI: 10.3390/microorganisms6040107] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
Abstract
'Leaky gut' syndrome, long-associated with celiac disease, has attracted much attention in recent years and for decades, was widely known in complementary/alternative medicine circles. It is often described as an increase in the permeability of the intestinal mucosa, which could allow bacteria, toxic digestive metabolites, bacterial toxins, and small molecules to 'leak' into the bloodstream. Nervous system involvement with celiac disease is know to occur even at subclinical levels. Gluten and gluten sensitivity are considered to trigger this syndrome in individuals genetically predisposed to celiac disease. However, the incidence of celiac disease in the general population is quite low. Nevertheless, increased public interest in gluten sensitivity has contributed to expanded food labels stating 'gluten-free' and the proliferation of gluten-free products, which further drives gluten-free lifestyle changes by individuals without frank celiac disease. Moreover, systemic inflammation is associated with celiac disease, depression, and psychiatric comorbidities. This mini-review focuses on the possible neurophysiological basis of leaky gut; leaky brain disease; and the microbiota's contribution to inflammation, gastrointestinal, and blood-brain barrier integrity, in order to build a case for possible mechanisms that could foster further 'leaky' syndromes. We ask whether a gluten-free diet is important for anyone or only those with celiac disease.
Collapse
Affiliation(s)
- Mark E M Obrenovich
- Research Service, Louis Stokes Cleveland Department of Veteran's Affairs Medical Center, Cleveland, OH 44106, USA.
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA.
- The Gilgamesh Foundation for Medical Science and Research, Cleveland, OH 44116, USA.
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA.
- Departments of Chemistry and Biological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA.
| |
Collapse
|
44
|
The neuroprotective role of the brain opioid system in stroke injury. Drug Discov Today 2018; 23:1385-1395. [DOI: 10.1016/j.drudis.2018.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 11/18/2022]
|
45
|
Durrant A, Swift M, Beazley-Long N. A role for pericytes in chronic pain? Curr Opin Support Palliat Care 2018; 12:154-161. [PMID: 29553988 PMCID: PMC6027993 DOI: 10.1097/spc.0000000000000342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE OF REVIEW The importance of the blood-brain barrier (BBB) and neuroinflammation in neurodegenerative conditions is becoming increasingly apparent, yet very little is known about these neurovascular functions in nonmalignant disease chronic pain. Neural tissue pericytes play critical roles in the formation and maintenance of the BBB. Herein, we review the important roles of neural pericytes and address their potential role in chronic pain. RECENT FINDINGS Pericytes are implicated in the function of neural microvasculature, including BBB permeability, neuroimmune factor secretion and leukocyte transmigration. In addition, the multipotent stem cell nature of pericytes affords pericytes the ability to migrate into neural parenchyma and differentiate into pain-associated cell types. These recent findings indicate that pericytes are key players in pathological BBB disruption and neuroinflammation, and as such pericytes may be key players in chronic pain states. SUMMARY Pericytes play key roles in pathological processes associated with chronic pain. We propose that pericytes may be a therapeutic target for painful diseases that have associated neural vascular dysfunction. Given the paucity of new pharmacotherapies for chronic pain conditions, we hope that this review inspires researchers to unearth the potential role(s) of pericytes in chronic pain sowing the seeds for future new chronic pain therapies.
Collapse
Affiliation(s)
- A.M. Durrant
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH
| | - M.N Swift
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH
| | - N. Beazley-Long
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH
| |
Collapse
|
46
|
Beazley-Long N, Durrant AM, Swift MN, Donaldson LF. The physiological functions of central nervous system pericytes and a potential role in pain. F1000Res 2018; 7:341. [PMID: 29623199 PMCID: PMC5861511 DOI: 10.12688/f1000research.13548.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2018] [Indexed: 12/29/2022] Open
Abstract
Central nervous system (CNS) pericytes regulate critical functions of the neurovascular unit in health and disease. CNS pericytes are an attractive pharmacological target for their position within the neurovasculature and for their role in neuroinflammation. Whether the function of CNS pericytes also affects pain states and nociceptive mechanisms is currently not understood. Could it be that pericytes hold the key to pain associated with CNS blood vessel dysfunction? This article reviews recent findings on the important physiological functions of CNS pericytes and highlights how these neurovascular functions could be linked to pain states.
Collapse
Affiliation(s)
- Nicholas Beazley-Long
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Alexandra M Durrant
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Matthew N Swift
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Lucy F Donaldson
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| |
Collapse
|
47
|
Dadas A, Washington J, Diaz-Arrastia R, Janigro D. Biomarkers in traumatic brain injury (TBI): a review. Neuropsychiatr Dis Treat 2018; 14:2989-3000. [PMID: 30510421 PMCID: PMC6231511 DOI: 10.2147/ndt.s125620] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Biomarkers can be broadly defined as qualitative or quantitative measurements that convey information on the physiopathological state of a subject at a certain time point or disease state. Biomarkers can indicate health, pathology, or response to treatment, including unwanted side effects. When used as outcomes in clinical trials, biomarkers act as surrogates or substitutes for clinically meaningful endpoints. Biomarkers of disease can be diagnostic (the identification of the nature and cause of a condition) or prognostic (predicting the likelihood of a person's survival or outcome of a disease). In addition, genetic biomarkers can be used to quantify the risk of developing a certain disease. In the specific case of traumatic brain injury, surrogate blood biomarkers of imaging can improve the standard of care and reduce the costs of diagnosis. In addition, a prognostic role for biomarkers has been suggested in the case of post-traumatic epilepsy. Given the extensive literature on clinical biomarkers, we will focus herein on biomarkers which are present in peripheral body fluids such as saliva and blood. In particular, blood biomarkers, such as glial fibrillary acidic protein and salivary/blood S100B, will be discussed together with the use of nucleic acids (eg, DNA) collected from peripheral cells.
Collapse
Affiliation(s)
| | | | | | - Damir Janigro
- FloTBI Inc., Cleveland, OH, USA, .,Department of Physiology, Case Western Reserve University, Cleveland, OH, USA,
| |
Collapse
|
48
|
Association of inflammatory mediators with pain perception. Biomed Pharmacother 2017; 96:1445-1452. [DOI: 10.1016/j.biopha.2017.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 12/31/2022] Open
|
49
|
Dkhar LK, Bartley J, White D, Seyfoddin A. Intranasal drug delivery devices and interventions associated with post-operative endoscopic sinus surgery. Pharm Dev Technol 2017; 23:282-294. [DOI: 10.1080/10837450.2017.1389956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Lari K. Dkhar
- Drug Delivery Research Group, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Jim Bartley
- Bio Design Lab, School of Engineering, Auckland University of Technology, Auckland, New Zealand
- Counties Manukau District Health Board, Auckland, New Zealand
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - David White
- Counties Manukau District Health Board, Auckland, New Zealand
| | - Ali Seyfoddin
- Drug Delivery Research Group, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
- Bio Design Lab, School of Engineering, Auckland University of Technology, Auckland, New Zealand
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|