51
|
Effects of Docosahexaenoic Acid and Its Peroxidation Product on Amyloid-β Peptide-Stimulated Microglia. Mol Neurobiol 2019; 57:1085-1098. [PMID: 31677009 DOI: 10.1007/s12035-019-01805-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
Abstract
Growing evidence suggests that docosahexaenoic acid (DHA) exerts neuroprotective effects, although the mechanism(s) underlying these beneficial effects are not fully understood. Here we demonstrate that DHA, but not arachidonic acid (ARA), suppressed oligomeric amyloid-β peptide (oAβ)-induced reactive oxygen species (ROS) production in primary mouse microglia and immortalized mouse microglia (BV2). Similarly, DHA but not ARA suppressed oAβ-induced increases in phosphorylated cytosolic phospholipase A2 (p-cPLA2), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-α (TNF-α) in BV2 cells. LC-MS/MS assay indicated the ability for DHA to cause an increase in 4-hydroxyhexenal (4-HHE) and suppress oAβ-induced increase in 4-hydroxynonenal (4-HNE). Although oAβ did not alter the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, exogenous DHA, ARA as well as low concentrations of 4-HHE and 4-HNE upregulated this pathway and increased production of heme oxygenase-1 (HO-1) in microglial cells. These results suggest that DHA modulates ARA metabolism in oAβ-stimulated microglia through suppressing oxidative and inflammatory pathways and upregulating the antioxidative stress pathway involving Nrf2/HO-1. Understanding the mechanism(s) underlying the beneficial effects of DHA on microglia should shed light into nutraceutical therapy for the prevention and treatment of Alzheimer's disease (AD).
Collapse
|
52
|
Cai W, Dai X, Chen J, Zhao J, Xu M, Zhang L, Yang B, Zhang W, Rocha M, Nakao T, Kofler J, Shi Y, Stetler RA, Hu X, Chen J. STAT6/Arg1 promotes microglia/macrophage efferocytosis and inflammation resolution in stroke mice. JCI Insight 2019; 4:131355. [PMID: 31619589 PMCID: PMC6824303 DOI: 10.1172/jci.insight.131355] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Efferocytosis, or phagocytic clearance of dead/dying cells by brain-resident microglia and/or infiltrating macrophages, is instrumental for inflammation resolution and restoration of brain homeostasis after stroke. Here, we identify the signal transducer and activator of transcription 6/arginase1 (STAT6/Arg1) signaling axis as a potentially novel mechanism that orchestrates microglia/macrophage responses in the ischemic brain. Activation of STAT6 was observed in microglia/macrophages in the ischemic territory in a mouse model of stroke and in stroke patients. STAT6 deficiency resulted in reduced clearance of dead/dying neurons, increased inflammatory gene signature in microglia/macrophages, and enlarged infarct volume early after experimental stroke. All of these pathological changes culminated in an increased brain tissue loss and exacerbated long-term functional deficits. Combined in vivo analyses using BM chimeras and in vitro experiments using microglia/macrophage-neuron cocultures confirmed that STAT6 activation in both microglia and macrophages was essential for neuroprotection. Adoptive transfer of WT macrophages into STAT6-KO mice reduced accumulation of dead neurons in the ischemic territory and ameliorated brain infarction. Furthermore, decreased expression of Arg1 in STAT6-/- microglia/macrophages was responsible for impairments in efferocytosis and loss of antiinflammatory modality. Our study suggests that efferocytosis via STAT6/Arg1 modulates microglia/macrophage phenotype, accelerates inflammation resolution, and improves stroke outcomes.
Collapse
Affiliation(s)
- Wei Cai
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xuejiao Dai
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jie Chen
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Jingyan Zhao
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mingyue Xu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lili Zhang
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Boyu Yang
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wenting Zhang
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marcelo Rocha
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Toshimasa Nakao
- T.E. Starzl Transplantation Institute and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - R. Anne Stetler
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
53
|
Halstead MR, Geocadin RG. The Medical Management of Cerebral Edema: Past, Present, and Future Therapies. Neurotherapeutics 2019; 16:1133-1148. [PMID: 31512062 PMCID: PMC6985348 DOI: 10.1007/s13311-019-00779-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Cerebral edema is commonly associated with cerebral pathology, and the clinical manifestation is largely related to the underlying lesioned tissue. Brain edema usually amplifies the dysfunction of the lesioned tissue and the burden of cerebral edema correlates with increased morbidity and mortality across diseases. Our modern-day approach to the medical management of cerebral edema has largely revolved around, an increasingly artificial distinction between cytotoxic and vasogenic cerebral edema. These nontargeted interventions such as hyperosmolar agents and sedation have been the mainstay in clinical practice and offer noneloquent solutions to a dire problem. Our current understanding of the underlying molecular mechanisms driving cerebral edema is becoming much more advanced, with differences being identified across diseases and populations. As our understanding of the underlying molecular mechanisms in neuronal injury continues to expand, so too is the list of targeted therapies in the pipeline. Here we present a brief review of the molecular mechanisms driving cerebral edema and a current overview of our understanding of the molecular targets being investigated.
Collapse
Affiliation(s)
- Michael R Halstead
- Neurosciences Critical Care Division, Departments of Neurology, Anesthesiology-Critical Care Medicine and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA.
| | - Romergryko G Geocadin
- Neurosciences Critical Care Division, Departments of Neurology, Anesthesiology-Critical Care Medicine and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| |
Collapse
|
54
|
Saito M, Saito M, Das BC. Involvement of AMP-activated protein kinase in neuroinflammation and neurodegeneration in the adult and developing brain. Int J Dev Neurosci 2019; 77:48-59. [PMID: 30707928 PMCID: PMC6663660 DOI: 10.1016/j.ijdevneu.2019.01.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/29/2018] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Microglial activation followed by neuroinflammation is a defense mechanism of the brain to eliminate harmful endogenous and exogenous materials including pathogens and damaged tissues, while excessive or chronic neuroinflammation may cause or exacerbate neurodegeneration observed in brain injuries and neurodegenerative diseases. Depending on conditions/environments during activation, microglia acquire distinct phenotypes, such as pro-inflammatory, anti-inflammatory, and disease-associated phenotypes, and show their ability to phagocytose various objects and produce pro-and anti-inflammatory mediators. Prevention of excessive inflammation by regulating the microglia's pro/anti-inflammatory balance is important for alleviating progression of brain injuries and diseases. Among many factors involved in the regulation of microglial phenotypes, cellular energy status plays an important role. Adenosine monophosphate-activated protein kinase (AMPK), which serves as a master sensor and regulator of energy balance, is considered a candidate molecule. Accumulating evidence from adult rodent studies indicates that AMPK activation promotes anti-inflammatory responses in microglia exposed to danger signals or various stressors mainly through inhibition of the nuclear factor κB (NF-κB) signaling and activation of the nuclear factor erythroid-2-related factor-2 (Nrf2) pathway. However, AMPK activation in neurons exposed to stressors/insults may exacerbate neuronal damage if AMPK activation is excessive or prolonged. While AMPK affects microglial activation states and neuronal cell survival rates in both the adult and the developing brain, studies in the developing brain are still scarce, even though activated AMPK is highly expressed especially in the neonatal brain. More in depth studies in the developing brain are important, because neuroinflammation/neurodegeneration occurred during development can result in long-lasting brain damage.
Collapse
Affiliation(s)
- Mariko Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research 140 Old Orangeburg, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Langone Medical Center 550 First Avenue, New York, NY 10016, USA
| | - Mitsuo Saito
- Division of Analytical Psychopharmacology, Nathan S. Kline Institute for Psychiatric Research 140 Old Orangeburg, Orangeburg, NY 10962, USA
| | - Bhaskar C. Das
- Departments of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai 1468 Madison Avenue, Annenberg 19-201, New York, NY 10029, USA
| |
Collapse
|
55
|
Shao Z, Tu S, Shao A. Pathophysiological Mechanisms and Potential Therapeutic Targets in Intracerebral Hemorrhage. Front Pharmacol 2019; 10:1079. [PMID: 31607923 PMCID: PMC6761372 DOI: 10.3389/fphar.2019.01079] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of hemorrhagic stroke with high mortality and morbidity. The resulting hematoma within brain parenchyma induces a series of adverse events causing primary and secondary brain injury. The mechanism of injury after ICH is very complicated and has not yet been illuminated. This review discusses some key pathophysiology mechanisms in ICH such as oxidative stress (OS), inflammation, iron toxicity, and thrombin formation. The corresponding therapeutic targets and therapeutic strategies are also reviewed.
Collapse
Affiliation(s)
- Zhiwei Shao
- Department of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- Department of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
56
|
Grijalva-Avila J, Villanueva-Fierro I, Lares-Asseff I, Chairez-Hernández I, Rivera-Sanchez G, Martínez-Estrada S, Martínez-Rivera I, Quiñones LA, Loera-Castañeda V. Milk intake and IGF-1 rs6214 polymorphism as protective factors to obesity. Int J Food Sci Nutr 2019; 71:388-393. [DOI: 10.1080/09637486.2019.1666805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | - Ismael Lares-Asseff
- Instituto Politécnico, Nacional-CIIDIR Unidad Durango, Durango, México
- Latin-American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | | | - Gildardo Rivera-Sanchez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, México
| | | | | | - Luis A. Quiñones
- Latin-American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Verónica Loera-Castañeda
- Instituto Politécnico, Nacional-CIIDIR Unidad Durango, Durango, México
- Latin-American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| |
Collapse
|
57
|
Vockley J, Dobrowolski SF, Arnold GL, Guerrero RB, Derks TGJ, Weinstein DA. Complex patterns of inheritance, including synergistic heterozygosity, in inborn errors of metabolism: Implications for precision medicine driven diagnosis and treatment. Mol Genet Metab 2019; 128:1-9. [PMID: 31358473 PMCID: PMC8931500 DOI: 10.1016/j.ymgme.2019.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 01/03/2023]
Abstract
Inborn errors of metabolism have traditionally been viewed as the quintessential single gene disorders; defects in one gene leads to loss of activity of one enzyme causing a metabolic imbalance and clinical disease. However, reality has never been quite that simple, and the classic "one gene-one enzyme" paradigm has been upended in many ways. Multiple gene defects can lead to the same biochemical phenotype, often with different clinical symptoms. Additionally, different mutations in the same gene can cause variable phenotypes, often most dramatic when a disease can be identified by pre-symptomatic screening. Moreover, response to therapy is not homogeneous across diseases and specific mutations. Perhaps the biggest deviation from traditional monogenic inheritance is in the setting of synergistic heterozygosity, a multigenic inheritance pattern in which mutations in multiple genes in a metabolic pathway lead to sufficient disruption of flux through the pathway, mimicking a monogenic disorder caused by homozygous defects in one gene in that pathway. In addition, widespread adoption of whole exome and whole genome sequencing in medical genetics has led to the realization that individual patients with apparently hybrid phenotypes can have mutations in more than one gene, leading to a mixed genetic disorder. Each of these situations point to a need for as much precision as possible in diagnosing metabolic disease, and it is likely to become increasingly critical to drive therapy. This article examines examples in traditional monogenic disorders that illustrates these points and define inborn errors of metabolism as complex genetic traits on the leading edge of precision medicine.
Collapse
Affiliation(s)
- Jerry Vockley
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America.
| | - Steven F Dobrowolski
- University of Pittsburgh School of Medicine, Department of Pathology, Pittsburgh, PA. UPMC Children's Hospital of Pittsburgh. 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | - Georgianne L Arnold
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | | | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO box 30 001, 9700, RB, Groningen, the Netherlands
| | - David A Weinstein
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, United States of America; GSD Program, Connecticut Children's Medical Center, Hartford, CT 06106, United States of America
| |
Collapse
|
58
|
Stamova B, Ander BP, Jickling G, Hamade F, Durocher M, Zhan X, Liu DZ, Cheng X, Hull H, Yee A, Ng K, Shroff N, Sharp FR. The intracerebral hemorrhage blood transcriptome in humans differs from the ischemic stroke and vascular risk factor control blood transcriptomes. J Cereb Blood Flow Metab 2019; 39:1818-1835. [PMID: 29651892 PMCID: PMC6727143 DOI: 10.1177/0271678x18769513] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding how the blood transcriptome of human intracerebral hemorrhage (ICH) differs from ischemic stroke (IS) and matched controls (CTRL) will improve understanding of immune and coagulation pathways in both disorders. This study examined RNA from 99 human whole-blood samples using GeneChip® HTA 2.0 arrays to assess differentially expressed transcripts of alternatively spliced genes between ICH, IS and CTRL. We used a mixed regression model with FDR-corrected p(Dx) < 0.2 and p < 0.005 and |FC| > 1.2 for individual comparisons. For time-dependent analyses, subjects were divided into four time-points: 0(CTRL), <24 h, 24-48 h, >48 h; 489 transcripts were differentially expressed between ICH and CTRL, and 63 between IS and CTRL. ICH had differentially expressed T-cell receptor and CD36 genes, and iNOS, TLR, macrophage, and T-helper pathways. IS had more non-coding RNA. ICH and IS both had angiogenesis, CTLA4 in T lymphocytes, CD28 in T helper cells, NFAT regulation of immune response, and glucocorticoid receptor signaling pathways. Self-organizing maps revealed 4357 transcripts changing expression over time in ICH, and 1136 in IS. Understanding ICH and IS transcriptomes will be useful for biomarker development, treatment and prevention strategies, and for evaluating how well animal models recapitulate human ICH and IS.
Collapse
Affiliation(s)
- Boryana Stamova
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Bradley P Ander
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Glen Jickling
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA.,2 Department of Medicine, University of Alberta, Edmonton, Canada
| | - Farah Hamade
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Marc Durocher
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Xinhua Zhan
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Da Zhi Liu
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Xiyuan Cheng
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Heather Hull
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Alan Yee
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Kwan Ng
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Natasha Shroff
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Frank R Sharp
- 1 Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
59
|
Abstract
Haemoglobin is released into the CNS during the breakdown of red blood cells after intracranial bleeding. Extracellular free haemoglobin is directly neurotoxic. Haemoglobin scavenging mechanisms clear haemoglobin and reduce toxicity; these mechanisms include erythrophagocytosis, haptoglobin binding of haemoglobin, haemopexin binding of haem and haem oxygenase breakdown of haem. However, the capacity of these mechanisms is limited in the CNS, and they easily become overwhelmed. Targeting of haemoglobin toxicity and scavenging is, therefore, a rational therapeutic strategy. In this Review, we summarize the neurotoxic mechanisms of extracellular haemoglobin and the peculiarities of haemoglobin scavenging pathways in the brain. Evidence for a role of haemoglobin toxicity in neurological disorders is discussed, with a focus on subarachnoid haemorrhage and intracerebral haemorrhage, and emerging treatment strategies based on the molecular pathways involved are considered. By focusing on a fundamental biological commonality between diverse neurological conditions, we aim to encourage the application of knowledge of haemoglobin toxicity and scavenging across various conditions. We also hope that the principles highlighted will stimulate research to explore the potential of the pathways discussed. Finally, we present a consensus opinion on the research priorities that will help to bring about clinical benefits.
Collapse
|
60
|
Wang Z, Chen Z, Yang J, Yang Z, Yin J, Duan X, Shen H, Li H, Wang Z, Chen G. Treatment of secondary brain injury by perturbing postsynaptic density protein-95-NMDA receptor interaction after intracerebral hemorrhage in rats. J Cereb Blood Flow Metab 2019; 39. [PMID: 29513122 PMCID: PMC6681427 DOI: 10.1177/0271678x18762637] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Postsynaptic density protein-95 (PSD95) plays important roles in the formation, differentiation, remodeling, and maturation of neuronal synapses. This study is to estimate the potential role of PSD95 in cognitive dysfunction and synaptic injury following intracerebral hemorrhage (ICH). The interaction between PSD95 and NMDA receptor subunit NR2B-neurotransmitter nitric oxide synthase (nNOS) could form a signal protein complex mediating excitatory signaling. Besides NR2B-nNOS, PSD95 also can bind to neurexin-1-neuroligin-1 to form a complex and participates in maintaining synaptic function. In this study, we found that there were an increase in the formation of PSD95-NR2B-nNOS complex and a decrease in the formation of neurexin-1-neuroligin-1-PSD95 complex after ICH, and this was accompanied by increased neuronal death and degeneration, and behavior dysfunction. PSD95 inhibitor Tat-NR2B9c effectively inhibited the interaction between PSD95 and NR2B-nNOS, and promoted the formation of neurexin-1-nueuroligin-1-PSD95 complex. In addition, Tat-NR2B9c treatment significantly reduced neuronal death and degeneration and matrix metalloproteinase 9 activity, alleviated inflammatory response and neurobehavioral disorders, and improved the cognitive and learning ability of ICH rats. Inhibition of the formation of PSD95-NR2B-nNOS complex can rescue secondary brain injury and behavioral cognitive impairment after ICH. PSD95 is expected to be a target for improving the prognosis of patients with ICH.
Collapse
Affiliation(s)
- Zhifeng Wang
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhouqing Chen
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Junjie Yang
- 2 Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, China
| | - Ziying Yang
- 2 Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, China
| | - Jia Yin
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaochun Duan
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haitao Shen
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiying Li
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- 1 Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
61
|
Walsh KB, Zhang X, Zhu X, Wohleb E, Woo D, Lu L, Adeoye O. Intracerebral Hemorrhage Induces Inflammatory Gene Expression in Peripheral Blood: Global Transcriptional Profiling in Intracerebral Hemorrhage Patients. DNA Cell Biol 2019; 38:660-669. [PMID: 31120332 PMCID: PMC6909779 DOI: 10.1089/dna.2018.4550] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/03/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023] Open
Abstract
To perform global transcriptome profiling using RNA-seq in the peripheral blood of intracerebral hemorrhage (ICH) patients. In 11 patients with ICH, peripheral blood was collected within 24 h of symptom onset or last known well, and a second blood draw occurred 72 h (±6) after the first. RNA-seq identified differentially expressed genes (DEGs) between the first and second samples. Biological pathway enrichment analysis was performed with Ingenuity® Pathway Analysis (IPA). A total of 16,640 genes were identified and 218 were significant DEGs after ICH (false discovery rate <0.1). IPA identified 97 disease and functional categories that were significantly upregulated (z-score >2) post-ICH; 46 categories were specifically related to immune cell activation, 22 to general cellular activation processes, and 4 to other inflammation-related responses. In the canonical pathway and network analysis, inflammatory mediators of particular importance included interleukin-8, NF-κB, ERK1/2, and members of the integrin class. ICH induced peripheral blood gene expression at 72 to 96 h compared with 0 to 24 h from symptom onset. DEGs that were highly expressed included those related to inflammation and activation of the immune response. Further research is needed to determine whether these changes affect outcomes and may represent new therapeutic targets.
Collapse
Affiliation(s)
- Kyle B. Walsh
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, Ohio
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio
| | - Xiaoting Zhu
- Division of Biomedical Informatics, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Eric Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio
- University of Cincinnati Neurobiology Research Center, Cincinnati, Ohio
| | - Daniel Woo
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, Ohio
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Long Lu
- Division of Biomedical Informatics, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Opeolu Adeoye
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, Ohio
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
62
|
Xie YZ, Zhang XJ, Zhang C, Yang Y, He JN, Chen YX. Protective effects of leonurine against ischemic stroke in mice by activating nuclear factor erythroid 2-related factor 2 pathway. CNS Neurosci Ther 2019; 25:1006-1017. [PMID: 31087454 PMCID: PMC6698971 DOI: 10.1111/cns.13146] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Aims Leonurine has been shown to trigger antioxidant responses during ischemic stroke, and nuclear factor erythroid 2‐related factor 2 (Nrf‐2) imparts protective effects against oxidative injury. The present study has determined that leonurine prevents ischemic injury of brain tissues via Nrf‐2 pathway activation. Methods Male ICR mice and Nrf‐2−/− mice were subjected to permanent middle cerebral artery occlusion (pMCAO) and received leonurine treatment at 2 hours after pMCAO by intraperitoneal injection. Neurological deficit scores as well as infarct volume were assessed to determine the neuroprotective role of leonurine. Nrf‐2 was investigated using Western blotting and real‐time polymerase chain reaction (RT‐PCR) analysis to elucidate the neuroprotective mechanism of leonurine. Commercial kits were employed to determine reactive oxygen species (ROS), superoxide (SOD), catalase (CAT), glutathione peroxidase (GSH‐Px), malonaldehyde (MDA), and glutathione (GSH). Vascular endothelial growth factor (VEGF) was evaluated by Western blotting and RT‐PCR analysis, and VEGF was localized using immunofluorescence. Results The application of leonurine on ICR mice resulted in an improvement in neurological deficit scores and a reduction in infarct volume. Leonurine upregulated nuclear Nrf‐2 protein and increased total Nrf‐2 protein expression and mRNA levels. Leonurine regulated SOD, MDA, CAT, GSH, and GSH‐Px, and it significantly inhibited ROS production in ICR mice. Leonurine improved VEGF expression and increased VEGF expression in neurons, astrocytes, and endothelial cells. However, leonurine had no obvious beneficial effects on Nrf‐2−/− mice. Conclusions Leonurine exerted neuroprotective effects, promoted antioxidant responses, and upregulated VEGF expression by activating the Nrf‐2 pathway.
Collapse
Affiliation(s)
- Yan-Zhao Xie
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Jian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yang Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Jun-Na He
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yan-Xia Chen
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
63
|
Macáková K, Afonso R, Saso L, Mladěnka P. The influence of alkaloids on oxidative stress and related signaling pathways. Free Radic Biol Med 2019; 134:429-444. [PMID: 30703480 DOI: 10.1016/j.freeradbiomed.2019.01.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 12/19/2022]
Abstract
Alkaloids have always attracted scientific interest due to either their positive or negative effects on human beings. This review aims to summarize their antioxidant effects by both classical in vitro scavenging assay and at the cellular level. Since most in vitro studies used the DPPH (2,2-diphenyl-1-picrylhydrazyl) radical scavenging assay, the results from those studies are summed up in the first part of the article. In the second part, available data on the effect of alkaloids on NADPH-oxidase, the key enzyme for reactive oxygen species production, at the cellular level, are summarized. More than 130 alkaloids were tested by DPPH assay. However, due to methodological differences, a direct comparison is hardly possible. It can be at least concluded that some of them were either similar to or even more active than standard antioxidants and the number of aromatic hydroxyl groups seems to be the major determinant for the activity. The data on inhibition of NADPH-oxidase activity by alkaloids demonstrated that there is little relationship to the DPPH assay. The mechanism seems to be based on inhibition of synthesis, activation or translocation of NADPH-oxidase subunits. In some alkaloids, activation of the nuclear factor Nrf2 pathway was documented to be the grounds for inhibition of NADPH-oxidase. Interestingly, many alkaloids can behave both as anti-oxidants and pro-oxidants depending on conditions and pro-oxidation might be the reason for activation of Nrf2. Available data on other "antioxidant" transcription factors FOXOs and PPARs are also mentioned.
Collapse
Affiliation(s)
- Kateřina Macáková
- Department of Pharmaceutical Botany, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| | - Rita Afonso
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, Italy.
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
64
|
Durocher M, Ander BP, Jickling G, Hamade F, Hull H, Knepp B, Liu DZ, Zhan X, Tran A, Cheng X, Ng K, Yee A, Sharp FR, Stamova B. Inflammatory, regulatory, and autophagy co-expression modules and hub genes underlie the peripheral immune response to human intracerebral hemorrhage. J Neuroinflammation 2019; 16:56. [PMID: 30836997 PMCID: PMC6399982 DOI: 10.1186/s12974-019-1433-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/12/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) has a high morbidity and mortality. The peripheral immune system and cross-talk between peripheral blood and brain have been implicated in the ICH immune response. Thus, we delineated the gene networks associated with human ICH in the peripheral blood transcriptome. We also compared the differentially expressed genes in blood following ICH to a prior human study of perihematomal brain tissue. METHODS We performed peripheral blood whole-transcriptome analysis of ICH and matched vascular risk factor control subjects (n = 66). Gene co-expression network analysis identified groups of co-expressed genes (modules) associated with ICH and their most interconnected genes (hubs). Mixed-effects regression identified differentially expressed genes in ICH compared to controls. RESULTS Of seven ICH-associated modules, six were enriched with cell-specific genes: one neutrophil module, one neutrophil plus monocyte module, one T cell module, one Natural Killer cell module, and two erythroblast modules. The neutrophil/monocyte modules were enriched in inflammatory/immune pathways; the T cell module in T cell receptor signaling genes; and the Natural Killer cell module in genes regulating alternative splicing, epigenetic, and post-translational modifications. One erythroblast module was enriched in autophagy pathways implicated in experimental ICH, and NRF2 signaling implicated in hematoma clearance. Many hub genes or module members, such as IARS, mTOR, S1PR1, LCK, FYN, SKAP1, ITK, AMBRA1, NLRC4, IL6R, IL17RA, GAB2, MXD1, PIK3CD, NUMB, MAPK14, DDX24, EVL, TDP1, ATG3, WDFY3, GSK3B, STAT3, STX3, CSF3R, PIP4K2A, ANXA3, DGAT2, LRP10, FLOT2, ANK1, CR1, SLC4A1, and DYSF, have been implicated in neuroinflammation, cell death, transcriptional regulation, and some as experimental ICH therapeutic targets. Gene-level analysis revealed 1225 genes (FDR p < 0.05, fold-change > |1.2|) have altered expression in ICH in peripheral blood. There was significant overlap of the 1225 genes with dysregulated genes in human perihematomal brain tissue (p = 7 × 10-3). Overlapping genes were enriched for neutrophil-specific genes (p = 6.4 × 10-08) involved in interleukin, neuroinflammation, apoptosis, and PPAR signaling. CONCLUSIONS This study delineates key processes underlying ICH pathophysiology, complements experimental ICH findings, and the hub genes significantly expand the list of novel ICH therapeutic targets. The overlap between blood and brain gene responses underscores the importance of examining blood-brain interactions in human ICH.
Collapse
Affiliation(s)
- Marc Durocher
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Bradley P. Ander
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Glen Jickling
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Farah Hamade
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Heather Hull
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Bodie Knepp
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Da Zhi Liu
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Xinhua Zhan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Anh Tran
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Xiyuan Cheng
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Kwan Ng
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Alan Yee
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Frank R. Sharp
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Boryana Stamova
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
- MIND Institute Biosciences Building, 2805 50th Street, Sacramento, CA 95817 USA
| |
Collapse
|
65
|
Bimodal Distribution of Nuclear Factor-κB Activation and Expression of Subunits in Experimental Models of Intracerebral Hemorrhage In Vivo. J Stroke Cerebrovasc Dis 2019; 28:821-829. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.11.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/12/2018] [Accepted: 11/29/2018] [Indexed: 02/08/2023] Open
|
66
|
Zuo Y, Huang L, Enkhjargal B, Xu W, Umut O, Travis ZD, Zhang G, Tang J, Liu F, Zhang JH. Activation of retinoid X receptor by bexarotene attenuates neuroinflammation via PPARγ/SIRT6/FoxO3a pathway after subarachnoid hemorrhage in rats. J Neuroinflammation 2019; 16:47. [PMID: 30791908 PMCID: PMC6385420 DOI: 10.1186/s12974-019-1432-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/04/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a life-threatening subtype of stroke with high mortality and disabilities. Retinoid X receptor (RXR) has been shown to be neuroprotective against ischemia/reperfusion injury. This study aimed to investigate the effects of the selective RXR agonist bexarotene on neuroinflammation in a rat model of SAH. METHODS Two hundred male Sprague-Dawley rats were used. The endovascular perforation induced SAH. Bexarotene was administered intraperitoneally at 1 h after SAH induction. To investigate the underlying mechanism, the selective RXR antagonist UVI3003 and RXR siRNA or SIRT6 inhibitor OSS128167 was administered via intracerebroventricular 1 h before SAH induction. Post-SAH assessments including SAH grade, neurological score, brain water content, Western blot, and immunofluorescence were performed. RESULTS The endogenous RXR and sirtuin 6 (SIRT6) protein levels were increased after SAH. Bexarotene treatment significantly reduced brain edema and improved the short-/long-term neurological deficit after SAH. Mechanistically, bexarotene increased the levels of PPARγ and SIRT6; decreased the expression of phosphorylated FoxO3a (p-FoxO3a), IL-6, IL-1β, and TNF-a; and inhibited the microglia activation and neutrophils infiltration at 24 h after SAH. Either UVI3003, OSS128167, or RXR siRNA abolished the neuroprotective effects of bexarotene and its regulation on protein levels of PPARγ/SIRT6/p-FoxO3a after SAH. CONCLUSIONS The activation of RXR by bexarotene attenuated neuroinflammation and improved neurological deficits after SAH. The anti-neuroinflammatory effect was at least partially through regulating PPARγ/SIRT6/FoxO3a pathway. Bexarotene may be a promising therapeutic strategy in the management of SAH patients.
Collapse
Affiliation(s)
- Yuchun Zuo
- Department of Neurosurgery, Third XiangYa Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013 China
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
| | - Weilin Xu
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
| | - Ocak Umut
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
| | - Zachary D. Travis
- Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| | - Guangyu Zhang
- Mass Spectrometry Core Facility, Loma Linda University, Loma Linda, CA 92354 USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
| | - Fei Liu
- Department of Neurosurgery, Third XiangYa Hospital, Central South University, No.138 Tongzipo Road, Changsha, 410013 China
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St. Risley Hall, Loma Linda, CA 92354 USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA 92354 USA
| |
Collapse
|
67
|
Wang Y, Bao DJ, Xu B, Cheng CD, Dong YF, Wei XP, Niu CS. Neuroprotection mediated by the Wnt/Frizzled signaling pathway in early brain injury induced by subarachnoid hemorrhage. Neural Regen Res 2019; 14:1013-1024. [PMID: 30762013 PMCID: PMC6404485 DOI: 10.4103/1673-5374.250620] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The Wnt/Frizzled signaling pathway participates in many inflammation-linked diseases. However, the inflammatory response mediated by the Wnt/Frizzled signaling pathway in experimental subarachnoid hemorrhage has not been thoroughly investigated. Consequently, in this study, we examined the potential role of the Wnt/Frizzled signaling pathway in early brain injury in rat models of subarachnoid hemorrhage. Simultaneously, possible neuroprotective mechanisms were also investigated. Experimental subarachnoid hemorrhage rat models were induced by injecting autologous blood into the prechiasmatic cistern. Experiment 1 was designed to examine expression of the Wnt/Frizzled signaling pathway in early brain injury induced by subarachnoid hemorrhage. In total, 42 adult rats were divided into sham (injection of equivalent volume of saline), 6-, 12-, 24-, 48-, 72-hour, and 1-week subarachnoid hemorrhage groups. Experiment 2 was designed to examine neuroprotective mechanisms of the Wnt/Frizzled signaling pathway in early brain injury induced by subarachnoid hemorrhage. Rats were treated with recombinant human Wnt1 (rhwnt1), small interfering Wnt1 (siwnt1) RNA, and monoclonal antibody of Frizzled1 (anti-Frizzled1) at 48 hours after subarachnoid hemorrhage. Expression levels of Wnt1, Frizzled1, β-catenin, peroxisome proliferator-activated receptor-γ, CD36, and active nuclear factor-κB were examined by western blot assay and immunofluorescence staining. Microglia type conversion and inflammatory cytokine levels in brain tissue were examined by immunofluorescence staining and enzyme-linked immunosorbent assay. Our results show that compared with the sham group, expression levels of Wnt1, Frizzled1, and β-catenin were low and reduced to a minimum at 48 hours, gradually returning to baseline at 1 week after subarachnoid hemorrhage. rhwnt1 treatment markedly increased Wnt1 expression and alleviated subarachnoid hemorrhage-induced early brain injury (within 72 hours), including cortical cell apoptosis, brain edema, and neurobehavioral deficits, accompanied by increasing protein levels of β-catenin, CD36, and peroxisome proliferator-activated receptor-γ and decreasing protein levels of nuclear factor-κB. Of note, rhwnt1 promoted M2-type microglia conversion and inhibited release of inflammatory cytokines (interleukin-1β, interleukin-6, and tumor necrosis factor-α). In contrast, siwnt1 RNA and anti-Frizzled1 treatment both resulted in an opposite effect. In conclusion, the Wnt/Frizzled1 signaling pathway may participate in subarachnoid hemorrhage-induced early brain injury via inhibiting the inflammatory response, including regulating microglia type conversion and decreasing inflammatory cytokine release. The study was approved by the Animal Ethics Committee of Anhui Medical University and First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (approval No. LLSC-20180202) in May 2017.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurosurgery, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - De-Jun Bao
- Department of Neurosurgery, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Bin Xu
- Anhui Medical University Auhui Province Medical Genetic Center, Hefei, Anhui Province, China
| | - Chuan-Dong Cheng
- Department of Neurosurgery, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Yong-Fei Dong
- Department of Neurosurgery, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Xiang-Pin Wei
- Department of Neurosurgery, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Chao-Shi Niu
- Department of Neurosurgery, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China; Anhui Province Key Laboratory of Brain Function and Brain Disease, Hefei, Anhui Province, China
| |
Collapse
|
68
|
Bobinger T, Burkardt P, B Huttner H, Manaenko A. Programmed Cell Death after Intracerebral Hemorrhage. Curr Neuropharmacol 2018; 16:1267-1281. [PMID: 28571544 PMCID: PMC6251052 DOI: 10.2174/1570159x15666170602112851] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 06/01/2017] [Indexed: 01/01/2023] Open
Abstract
Background: Intracerebral hemorrhage (ICH) accounts for up to 15% of all strokes and is characterized by high rates of mortality and morbidity. The post-ICH brain injury can be distinguished in 1) primary, which are caused by disrup-tion and mechanical deformation of brain tissue due to hematoma growth and 2) secondary, which are induced by microglia activation, mitochondrial dysfunction, neurotransmitter and inflammatory mediator release. Although these events typically lead to necrosis, the occurrence of programmed cell death has also been reported after ICH. Methods: We reviewed recent publications describing advance in pre- and clinic ICH research. Results: At present, treatment of ICH patients is based on oral anticoagulant reversal, management of blood pressure and other medical complications. Several pre-clinical studies showed promising results and demonstrated that anti-oxidative and anti-inflammatory treatments reduced neuronal cell death, however, to date, all of these attempts have failed in randomized controlled clinical trials. Yet, the time frame of administration may be crucial in translation from animal to clinical studies. Furthermore, the latest pre-clinical research points toward the existence of other, apoptosis-unrelated forms kinds of pro-grammed cell death. Conclusion: Our review summarizes current knowledge of pathways leading to programmed cell death after ICH in addition to data from clinical trials. Some of the pre-clinical results have not yet demonstrated clinical confirmation, however they sig-nificantly contribute to our understanding of post-ICH pathology and can contribute to development of new therapeutic ap-proaches, decreasing mortality and improving ICH patients’ quality of life.
Collapse
Affiliation(s)
- Tobias Bobinger
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Petra Burkardt
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Hagen B Huttner
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| |
Collapse
|
69
|
Lee EJ, Park JS, Lee YY, Kim DY, Kang JL, Kim HS. Anti-inflammatory and anti-oxidant mechanisms of an MMP-8 inhibitor in lipoteichoic acid-stimulated rat primary astrocytes: involvement of NF-κB, Nrf2, and PPAR-γ signaling pathways. J Neuroinflammation 2018; 15:326. [PMID: 30470240 PMCID: PMC6260848 DOI: 10.1186/s12974-018-1363-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/07/2018] [Indexed: 01/06/2023] Open
Abstract
Background Recent evidence suggests that reactive astrocytes play an important role in neuroinflammation and neurodegenerative diseases. Thus, controlling astrocyte reactivity has been suggested as a promising strategy for treating neurodegenerative diseases. In the present study, we investigated whether a matrix metalloproteinase (MMP)-8 inhibitor, M8I, could control neuroinflammation in lipoteichoic acid (LTA)-stimulated rat primary astrocytes. Methods The effects of M8I on the expression of inducible nitric oxide synthase, cytokines, and MMPs were examined in LTA-stimulated rat primary astrocytes by ELISA, RT-PCR, and Western blot analysis. The effects of M8I on reactive oxygen species (ROS) generation and phase II antioxidant enzyme expression were examined by the DCF-DA assay, RT-PCR, and Western blot analysis. The detailed molecular mechanisms underlying the anti-inflammatory and antioxidant effects of M8I were analyzed by the electrophoretic mobility shift assay, the reporter gene assay, Western blot, and RT-PCR analysis. Results Treatment with LTA, a major cell wall component of Gram-positive bacteria, led to astrocyte activation and induced the expression of inflammatory molecules such as iNOS, COX-2, and pro-inflammatory cytokines. In addition, LTA induced the expression of MMPs such as MMP-1, MMP-3, MMP-8, MMP-9, and MMP-13 in rat primary astrocytes. Based on previous reports showing that MMP-8 plays a role as a proinflammatory mediator in microglia, we investigated whether MMP-8 is also involved in inflammatory reactions of reactive astrocytes. We found that treatment of astrocytes with M8I significantly inhibited LTA-induced expression of iNOS, TNF-α, IL-1β, IL-6, and TLR-2. In addition, M8I inhibited LTA-induced NF-κB, MAP kinase, and Akt activities, while it increased the anti-inflammatory PPAR-γ activities. Moreover, M8I showed antioxidant effects by suppressing ROS production in LTA- or H2O2-stimulated astrocytes. Interestingly, M8I increased the expression of phase II antioxidant enzymes such as hemeoxygenase-1, NQO1, catalase, and MnSOD by modulating the Nrf2/ARE signaling pathway. Conclusions The data collectively suggest the therapeutic potential of an MMP-8 inhibitor in neuroinflammatory disorders that are associated with astrocyte reactivity. Electronic supplementary material The online version of this article (10.1186/s12974-018-1363-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eun-Jung Lee
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Mok-6-dong 911-1, Yangchun-Ku, Seoul, 158-710, South Korea
| | - Jin-Sun Park
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Mok-6-dong 911-1, Yangchun-Ku, Seoul, 158-710, South Korea
| | - Yu-Young Lee
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Mok-6-dong 911-1, Yangchun-Ku, Seoul, 158-710, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Mok-6-dong 911-1, Yangchun-Ku, Seoul, 158-710, South Korea
| | - Jihee Lee Kang
- Department of Physiology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Mok-6-dong 911-1, Yangchun-Ku, Seoul, 158-710, South Korea.
| |
Collapse
|
70
|
Zhang W, Chen R, Yang T, Xu N, Chen J, Gao Y, Stetler RA. Fatty acid transporting proteins: Roles in brain development, aging, and stroke. Prostaglandins Leukot Essent Fatty Acids 2018; 136:35-45. [PMID: 28457600 PMCID: PMC5650946 DOI: 10.1016/j.plefa.2017.04.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022]
Abstract
Polyunsaturated fatty acids are required for the brain development and significantly impact aging and stroke. Due to the hydrophobicity of fatty acids, fatty acids transportation related proteins that include fatty acid binding proteins (FABPs), long chain acyl-coA synthase (ACS), fatty acid transportation proteins (FATPs), fatty acid translocase (FAT/CD36) and newly reported major facilitator superfamily domain-containing protein (Mfsd2a) play critical roles in the uptake of various fatty acids, especially polyunsaturated fatty acids. They are not only involved in neurodevelopment, but also have great impact on neurological disease, such as aging related dementia and stroke.
Collapse
Affiliation(s)
- Wenting Zhang
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ruiying Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Na Xu
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - R Anne Stetler
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA.
| |
Collapse
|
71
|
Lecca D, Janda E, Mulas G, Diana A, Martino C, Angius F, Spolitu S, Casu MA, Simbula G, Boi L, Batetta B, Spiga S, Carta AR. Boosting phagocytosis and anti-inflammatory phenotype in microglia mediates neuroprotection by PPARγ agonist MDG548 in Parkinson's disease models. Br J Pharmacol 2018; 175:3298-3314. [PMID: 29570770 DOI: 10.1111/bph.14214] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 03/02/2018] [Accepted: 03/04/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Microglial phenotype and phagocytic activity are deregulated in Parkinson's disease (PD). PPARγ agonists are neuroprotective in experimental PD, but their role in regulating microglial phenotype and phagocytosis has been poorly investigated. We addressed it by using the PPARγ agonist MDG548. EXPERIMENTAL APPROACH Murine microglial cell line MMGT12 was stimulated with LPS and/or MDG548, and their effect on phagocytosis of fluorescent microspheres or necrotic neurons was investigated by flow cytometry. Cytokines and markers of microglia phenotype, such as mannose receptor C type 1; MRC1), Ym1 and CD68 were measured by elisa and fluorescent immunohistochemistry. Levels of Beclin-1, which plays a role in microglial phagocytosis, were measured by Western blotting. In the in vivo MPTP-probenecid (MPTPp) model of PD in mice, MDG548 was tested on motor impairment, nigral neurodegeneration, microglial activation and phenotype. KEY RESULTS In LPS-stimulated microglia, MDG548 increased phagocytosis of both latex beads and necrotic cells, up-regulated the expression of MRC1, CD68 and to a lesser extent IL-10, while blocking the LPS-induced increase of TNF-α and iNOS. MDG548 also induced Beclin-1. Chronic MPTPp treatment in mice down-regulated MRC1 and TGF-β and up-regulated TNF-α and IL-1β immunoreactivity in activated CD11b-positive microglia, causing the death of nigral dopaminergic neurons. MDG548 arrested MPTPp-induced cell death, enhanced MRC1 and restored cytokine levels. CONCLUSIONS AND IMPLICATIONS This study adds a novel mechanism for PPARγ-mediated neuroprotection in PD and suggests that increasing phagocytic activity and anti-inflammatory markers may represent an effective disease-modifying approach.
Collapse
Affiliation(s)
- Daniela Lecca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Giovanna Mulas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Concetta Martino
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Fabrizio Angius
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Spolitu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Gabriella Simbula
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Laura Boi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Barbara Batetta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
72
|
PPAR- γ Promotes Hematoma Clearance through Haptoglobin-Hemoglobin-CD163 in a Rat Model of Intracerebral Hemorrhage. Behav Neurol 2018; 2018:7646104. [PMID: 30123388 PMCID: PMC6079602 DOI: 10.1155/2018/7646104] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/30/2022] Open
Abstract
Background and Purpose PPAR-γ is a transcriptional factor which is associated with promoting hematoma clearance and reducing neurological dysfunction after intracerebral hemorrhage (ICH). Haptoglobin- (Hp-) hemoglobin- (Hb-) CD163 acts as a main pathway to Hb scavenging after ICH. The effect of PPAR-γ on the Hp-Hb-CD163 signaling pathway has not been reported. We hypothesized that PPAR-γ might protect against ICH-induced neuronal injury via activating the Hp-Hb-CD163 pathway in a rat ICH model. Methods 107 Sprague-Dawley rats were used in this research. They were randomly allocated to 4 groups as follows: sham group, vehicle group, monascin-treated group, and Glivec-treated group. Animals were euthanized at 3 days after the model was established successfully. We observed the effects of PPAR-γ on the brain water content, hemoglobin levels, and the expressions of CD163 and Hp in Western blot and real-ime PCR; meanwhile, we measured hematoma volumes and edema areas by MRI scanning. Result The results showed that PPAR-γ agonist significantly reduced hematoma volume, brain edema, and hemoglobin after ICH. It also enhanced CD163 and Hp expression while PPAR-γ antagonist had the opposite effects. Conclusions PPAR-γ promotes hematoma clearance and plays a protective role through the Hp-Hb-CD163 pathway in a rat collagenase infusion ICH model.
Collapse
|
73
|
Combined Rosiglitazone and Forskolin Have Neuroprotective Effects in SD Rats after Spinal Cord Injury. PPAR Res 2018; 2018:3897478. [PMID: 30034460 PMCID: PMC6032969 DOI: 10.1155/2018/3897478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/22/2018] [Accepted: 05/08/2018] [Indexed: 02/05/2023] Open
Abstract
The peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist rosiglitazone inhibits NF-κB expression and endogenous neural stem cell differentiation into neurons and reduces the inflammatory cascade after spinal cord injury (SCI). The aim of this study was to explore the mechanisms underlying rosiglitazone-mediated neuroprotective effects and regulation of the balance between the inflammatory cascade and generation of endogenous spinal cord neurons by using a spinal cord-derived neural stem cell culture system as well as SD rat SCI model. Activation of PPAR-γ could promote neural stem cell proliferation and inhibit PKA expression and neuronal formation in vitro. In the SD rat SCI model, the rosiglitazone + forskolin group showed better locomotor recovery compared to the rosiglitazone and forskolin groups. MAP2 expression was higher in the rosiglitazone + forskolin group than in the rosiglitazone group, NF-κB expression was lower in the rosiglitazone + forskolin group than in the forskolin group, and NeuN expression was higher in the rosiglitazone + forskolin group than in the forskolin group. PPAR-γ activation likely inhibits NF-κB, thereby reducing the inflammatory cascade, and PKA activation likely promotes neuronal cell regeneration.
Collapse
|
74
|
Belém-Filho IJA, Ribera PC, Nascimento AL, Gomes ARQ, Lima RR, Crespo-Lopez ME, Monteiro MC, Fontes-Júnior EA, Lima MO, Maia CSF. Low doses of methylmercury intoxication solely or associated to ethanol binge drinking induce psychiatric-like disorders in adolescent female rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:184-194. [PMID: 29734102 DOI: 10.1016/j.etap.2018.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 06/08/2023]
Abstract
Methylmercury (MeHg) is an environmental contaminant that provokes damage to developing brain. Simultaneously, the consumption of ethanol among adolescents has increased. Evidence concerning the effects of MeHg low doses per se or associated with ethanol during adolescence are scarce. Thus, we investigate behavioral disorders resulted from exposure to MeHg low doses and co-intoxicated with ethanol in adolescent rats. Wistar rats received chronic exposure to low doses of MeHg (40 μg/kg/day for 5 weeks) and/or ethanol binge drinking (3 g/kg/day at 3 days per week for 5 weeks). Animals were submitted to behavioral assays to assess emotionality and cognitive function. Total mercury content was evaluated in the brain and hair. Oxidative parameters were analyzed in blood samples. MeHg at low doses or associated to ethanol binge drinking produced psychiatric-like disorders and cognitive impairment. Peripherally, MeHg altered oxidative parameters when associated to ethanol. Ethanol administration reduced brain mercury deposit. We proposed that ethanol reduces the necessity of mercury tissue levels to display psychiatric-like disorders/cognitive impairment.
Collapse
Affiliation(s)
| | - Paula Cardoso Ribera
- Laboratório de Farmacologia da Inflamação e Comportamento, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Aline Lima Nascimento
- Laboratório de Farmacologia da Inflamação e Comportamento, Universidade Federal do Pará, Belém, Pará, Brazil
| | | | - Rafael Rodrigues Lima
- Laboratório de Biologia Funcional e Estrutural, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Ensaios In Vitro, Imunologia e Microbiologia, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Marta Chagas Monteiro
- Laboratório de Farmacologia Molecular, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Enéas Andrade Fontes-Júnior
- Laboratório de Farmacologia da Inflamação e Comportamento, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Marcelo Oliveira Lima
- Laboratório de Toxicologia, Seção de Meio Ambiente, Instituto Evandro Chagas, Belém, Pará, Brazil
| | | |
Collapse
|
75
|
Xie Z, Huang L, Enkhjargal B, Reis C, Wan W, Tang J, Cheng Y, Zhang JH. Recombinant Netrin-1 binding UNC5B receptor attenuates neuroinflammation and brain injury via PPARγ/NFκB signaling pathway after subarachnoid hemorrhage in rats. Brain Behav Immun 2018; 69:190-202. [PMID: 29162556 PMCID: PMC5894358 DOI: 10.1016/j.bbi.2017.11.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/09/2017] [Accepted: 11/16/2017] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation is an essential mechanism involved in the pathogenesis of subarachnoid hemorrhage (SAH)-induced brain injury. Recently, Netrin-1 (NTN-1) is well established to exert anti-inflammatory property in non-nervous system diseases through inhibiting infiltration of neutrophil. The present study was designed to investigate the effects of NTN-1 on neuroinflammation, and the potential mechanism in a rat model of SAH. Two hundred and ninety-four male Sprague Dawley rats (weight 280-330 g) were subjected to the endovascular perforation model of SAH. Recombinant human NTN-1 (rh-NTN-1) was administered intravenously. Small interfering RNA (siRNA) of NTN-1 and UNC5B, and a selective PPARγ antagonist bisphenol A diglycidyl ether (BADGE) were applied. Post-SAH evaluations included neurobehavioral function, brain water content, Western blot analysis, and immunohistochemistry. Our results showed that endogenous NTN-1 and its receptor UNC5B level were increased after SAH. Administration of rh-NTN-1 reduced brain edema, ameliorated neurological impairments, and suppressed microglia activation after SAH, which were concomitant with PPARγ activation, inhibition of NFκB, and decrease in TNF-α, IL-6, and ICAM-1, as well as myeloperoxidase (MPO). Knockdown of endogenous NTN-1 increased expression of pro-inflammatory mediators and MPO, and aggravated neuroinflammation and brain edema. Moreover, knockdown of UNC5B using specific siRNA and inhibition of PPARγ with BADGE blocked the protective effects of rh-NTN-1. In conclusion, our findings indicated that exogenous rh-NTN-1 treatment attenuated neuroinflammation and neurological impairments through inhibiting microglia activation after SAH in rats, which is possibly mediated by UNC5B/PPARγ/NFκB signaling pathway. Exogenous NTN-1 may be a novel therapeutic agent to ameliorating early brain injury via its anti-inflammation effect.
Collapse
Affiliation(s)
- Zongyi Xie
- Department of Neurosurgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China,Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Cesar Reis
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Weifeng Wan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.
| | - John H. Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA,Corresponding author: John H. Zhang, MD, PhD, Departments of Anesthesiology, Physiology and Pharmacology and Neurosurgery, Loma Linda University School of Medicine, 11041 Campus St, Risley Hall, Room 219, Loma Linda, CA 92354, USA. Tel: 909-558-4723; Fax: 909-558-0119; , Yuan Cheng, MD, Department of Neurosurgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China. Tel: +8623-63693539; Fax: +8623-63693871;
| |
Collapse
|
76
|
Wang G, Wang L, Sun XG, Tang J. Haematoma scavenging in intracerebral haemorrhage: from mechanisms to the clinic. J Cell Mol Med 2017; 22:768-777. [PMID: 29278306 PMCID: PMC5783832 DOI: 10.1111/jcmm.13441] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 09/14/2017] [Indexed: 01/22/2023] Open
Abstract
The products of erythrocyte lyses, haemoglobin (Hb) and haem, are recognized as neurotoxins and the main contributors to delayed cerebral oedema and tissue damage after intracerebral haemorrhage (ICH). Finding a means to efficiently promote absorption of the haemolytic products (Hb and haem) around the bleeding area in the brain through stimulating the function of the body's own garbage cleaning system is a novel clinical challenge and critical for functional recovery after ICH. In this review, available information of the brain injury mechanisms underlying ICH and endogenous haematoma scavenging system is provided. Meanwhile, potential intervention strategies are discussed. Intracerebral blood itself has ‘toxic’ effects beyond its volume effect after ICH. Haptoglobin–Hb–CD163 as well as haemopexin–haem–LRP1 is believed to be the most important endogenous scavenging pathway which participates in blood components resolution following ICH. PPARγ–Nrf2 activates the aforementioned clearance pathway and then accelerates haematoma clearance. Meanwhile, the scavenger receptors as novel targets for therapeutic interventions to treat ICH are also highlighted.
Collapse
Affiliation(s)
- Gaiqing Wang
- The second Hospital of Shanxi Medical University, Tai Yuan, China
| | - Li Wang
- The second Hospital of Shanxi Medical University, Tai Yuan, China
| | - Xin-Gang Sun
- The second Hospital of Shanxi Medical University, Tai Yuan, China
| | - Jiping Tang
- Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
77
|
Neuroprotective Potential of Gentongping in Rat Model of Cervical Spondylotic Radiculopathy Targeting PPAR- γ Pathway. J Immunol Res 2017; 2017:9152960. [PMID: 29230425 PMCID: PMC5694586 DOI: 10.1155/2017/9152960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/15/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022] Open
Abstract
Cervical spondylotic radiculopathy (CSR) is the most general form of spinal degenerative disease and is characterized by pain and numbness of the neck and arm. Gentongping (GTP) granule, as a classical Chinese patent medicine, has been widely used in curing CSR, whereas the underlying mechanism remains unclear. Therefore, the aim of this study is to explore the pharmacological mechanisms of GTP on CSR. The rat model of CSR was induced by spinal cord injury (SCI). Our results showed that GTP could significantly alleviate spontaneous pain as well as ameliorate gait. The HE staining and Western blot results showed that GTP could increase the quantity of motoneuron and enhance the activation of peroxisome proliferator-activated receptor gamma (PPAR-γ) in the spinal cord tissues. Meanwhile, immunofluorescence staining analysis indicated that GTP could reduce the expression of TNF-α in the spinal cord tissues. Furthermore, the protein level of Bax was decreased whereas the protein levels of Bcl-2 and NF200 were increased after the GTP treatment. These findings demonstrated that GTP might modulate the PPAR-γ pathway by inhibiting the inflammatory response and apoptosis as well as by protecting the cytoskeletal integrity of the spinal cord, ultimately play a neuroprotective role in CSR.
Collapse
|
78
|
Choi MJ, Lee EJ, Park JS, Kim SN, Park EM, Kim HS. Anti-inflammatory mechanism of galangin in lipopolysaccharide-stimulated microglia: Critical role of PPAR-γ signaling pathway. Biochem Pharmacol 2017; 144:120-131. [DOI: 10.1016/j.bcp.2017.07.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022]
|
79
|
Choi MJ, Park JS, Park JE, Kim HS, Kim HS. Galangin Suppresses Pro-Inflammatory Gene Expression in Polyinosinic-Polycytidylic Acid-Stimulated Microglial Cells. Biomol Ther (Seoul) 2017; 25:641-647. [PMID: 29081092 PMCID: PMC5685434 DOI: 10.4062/biomolther.2017.173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 11/17/2022] Open
Abstract
Galangin (3,5,7-trihydroxyflavone) is a polyphenolic compound abundant in honey and medicinal herbs, such as Alpinia officinarum. In this study, we investigated the anti-inflammatory effects of galangin under in vitro and in vivo neuroinflammatory conditions caused by polyinosinic-polycytidylic acid (poly(I:C)), a viral mimic dsRNA analog. Galangin suppressed the production of nitric oxide, reactive oxygen species, and pro-inflammatory cytokines in poly(I:C)-stimulated BV2 microglia. On the other hand, galangin enhanced anti-inflammatory interleukin (IL)-10 production. Galangin also suppressed the expression of pro-inflammatory markers in poly(I:C)-injected mouse brains. Further mechanistic studies showed that galangin inhibited poly(I:C)-induced nuclear factor (NF)-κB activity and phosphorylation of Akt without affecting MAP kinases. Interestingly, galangin increased the expression and transcriptional activity of peroxisome proliferator-activated receptor (PPAR)-γ, known to play an anti-inflammatory role. To investigate whether PPAR-γ is involved in the anti-inflammatory function of galangin, BV2 cells were pre-treated with PPAR-γ antagonist before treatment of galangin. We found that PPAR-γ antagonist significantly blocked galangin-mediated upregulation of IL-10 and attenuated the inhibition of tumor necrosis factor (TNF)-α and IL-6 in poly(I:C)-stimulated microglia. In conclusion, our data suggest that PI3K/Akt, NF-κB, and PPAR-γ play a pivotal role in mediating the anti-inflammatory effects of galangin in poly(I:C)-stimulated microglia.
Collapse
Affiliation(s)
- Min-Ji Choi
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Jin-Sun Park
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Jung-Eun Park
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Han Su Kim
- Department of Otorhinolaryngology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| |
Collapse
|
80
|
Wang Y, Chen Q, Tan Q, Feng Z, He Z, Tang J, Feng H, Zhu G, Chen Z. Simvastatin accelerates hematoma resolution after intracerebral hemorrhage in a PPARγ-dependent manner. Neuropharmacology 2017; 128:244-254. [PMID: 29054366 DOI: 10.1016/j.neuropharm.2017.10.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/19/2017] [Accepted: 10/16/2017] [Indexed: 12/24/2022]
Abstract
To date, the neuroprotective effects of statins on intracerebral hemorrhage (ICH) are not well established. This study explored the effect and potential mechanism of simvastatin treatment on ICH. In the present study, the effects of simvastatin on hematoma absorption, neurological outcome, CD36 expression and microglia polarization were examined in rat model of ICH model. In the meantime, inhibitory effect of PPARγ inhibitor GW9662 was investigated following ICH. Additionally, the effect of simvastatin on PPARγ activation was also investigated in rat ICH model and primary microglia culture. Much more, the role of PPARγ and CD36 in simvastatin-mediated erythrocyte phagocytosis was also detected by using in vivo or in vitro phagocytosis models, respectively. After ICH, simvastatin promoted hematoma absorption and improved neurological outcome after ICH while upregulating CD36 expression and facilitating M2 phenotype polarization in perihematomal microglia. In addition, simvastatin increased PPARγ activation and reinforced microglia-induced erythrocyte phagocytosis in vivo and in vitro. All above effects of simvastatin were abolished by PPARγ inhibitor GW9662. In conclusion, our data suggested that simvastatin could enhance hematoma clearance and attenuate neurological deficits possibly by activating PPARγ.
Collapse
Affiliation(s)
- Yuelong Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Zhou Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Zhenlin He
- Department of Reproduction and Genetics, Reproductive Medical Centre, The First People's Hospital of Yunnan Province, 650200, China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
81
|
Cai W, Yang T, Liu H, Han L, Zhang K, Hu X, Zhang X, Yin KJ, Gao Y, Bennett MVL, Leak RK, Chen J. Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair. Prog Neurobiol 2017; 163-164:27-58. [PMID: 29032144 DOI: 10.1016/j.pneurobio.2017.10.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/06/2017] [Accepted: 10/08/2017] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a widely expressed ligand-modulated transcription factor that governs the expression of genes involved in inflammation, redox equilibrium, trophic factor production, insulin sensitivity, and the metabolism of lipids and glucose. Synthetic PPARγ agonists (e.g. thiazolidinediones) are used to treat Type II diabetes and have the potential to limit the risk of developing brain injuries such as stroke by mitigating the influence of comorbidities. If brain injury develops, PPARγ serves as a master gatekeeper of cytoprotective stress responses, improving the chances of cellular survival and recovery of homeostatic equilibrium. In the acute injury phase, PPARγ directly restricts tissue damage by inhibiting the NFκB pathway to mitigate inflammation and stimulating the Nrf2/ARE axis to neutralize oxidative stress. During the chronic phase of acute brain injuries, PPARγ activation in injured cells culminates in the repair of gray and white matter, preservation of the blood-brain barrier, reconstruction of the neurovascular unit, resolution of inflammation, and long-term functional recovery. Thus, PPARγ lies at the apex of cell fate decisions and exerts profound effects on the chronic progression of acute injury conditions. Here, we review the therapeutic potential of PPARγ in stroke and brain trauma and highlight the novel role of PPARγ in long-term tissue repair. We describe its structure and function and identify the genes that it targets. PPARγ regulation of inflammation, metabolism, cell fate (proliferation/differentiation/maturation/survival), and many other processes also has relevance to other neurological diseases. Therefore, PPARγ is an attractive target for therapies against a number of progressive neurological disorders.
Collapse
Affiliation(s)
- Wei Cai
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huan Liu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lijuan Han
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kai Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA
| | - Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA.
| |
Collapse
|
82
|
Cheng MC, Pan TM. Glyceryl 1,3-Dipalmitate Produced from Lactobacillus paracasei subspecies. paracasei NTU 101 Inhibits Oxygen-Glucose Deprivation and Reperfusion-Induced Oxidative Stress via Upregulation of Peroxisome Proliferator-Activated Receptor γ in Neuronal SH-SY5Y Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7926-7933. [PMID: 28829589 DOI: 10.1021/acs.jafc.7b02728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Glyceryl 1,3-dipalmitate (GD) purified from Lactobacillus paracasei subsp. paracasei NTU 101-fermented products has been demonstrated to possess neuroprotective properties. We determined the effect of GD on oxygen-glucose deprivation and reperfusion (OGD/R)-induced SH-SY5Y neuroblastoma cell death. GD ameliorated OGD/R-induced apoptosis by elevating the protein expression of nuclear peroxisome proliferator-activated receptor γ (PPARγ) and nuclear factor erythroid 2-related factor 2 (Nrf2), thereby attenuating reactive oxygen species (ROS) generation. Pretreatment with GD reduced nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) expression from 1.54 ± 0.27 to 0.84 ± 0.46, thereby attenuating the induction of pro-inflammatory mediators, and increased the plasma membrane Ca2+ ATPase (PMCA) levels from 0.81 ± 0.02 to 1.08 ± 0.06, thus reducing the levels of cytosolic Ca2+; this also correlated with reduced cell death. We conclude that GD prevents SH-SY5Y cells from injury after OGD/R insult, possibly by modulating oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Meng-Chun Cheng
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Number 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Tzu-Ming Pan
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Number 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
| |
Collapse
|
83
|
Wowk S, Fagan KJ, Ma Y, Nichol H, Colbourne F. Examining potential side effects of therapeutic hypothermia in experimental intracerebral hemorrhage. J Cereb Blood Flow Metab 2017; 37:2975-2986. [PMID: 27899766 PMCID: PMC5536807 DOI: 10.1177/0271678x16681312] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 11/16/2022]
Abstract
Studies treating intracerebral hemorrhage (ICH) with therapeutic hypothermia (TH) have shown inconsistent benefits. We hypothesized that TH's anti-inflammatory effects may be responsible as inflammatory cells are essential for removing degrading erythrocytes. Here, we subjected rats to a collagenase-induced striatal ICH followed by whole-body TH (∼33℃ for 11-72 h) or normothermia. We used X-ray fluorescence imaging to spatially quantify total and peri-hematoma iron three days post-injury. At three and seven days, we measured non-heme iron levels. Finally, hematoma volume was quantified on one, three, and seven days. In the injured hemisphere, total iron levels were elevated ( p < 0.001) with iron increasing in the peri-hematoma region ( p = 0.007). Non-heme iron increased from three to seven days (p < 0.001). TH had no effect on any measure of iron ( p ≥ 0.479). At one and three days, TH did not affect hematoma volume ( p ≥ 0.264); however, at seven days there was a four-fold increase in hematoma volume in 40% of treated animals ( p = 0.032). Thus, even when TH does not interfere with initial increases in total and non-heme iron or its containment, TH can cause re-bleeding post-treatment. This serious complication could partly account for the intermittent protection previously observed. This also raises serious concerns for clinical usage of TH for ICH.
Collapse
Affiliation(s)
- Shannon Wowk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kelly J Fagan
- Department of Biology, MacEwan University, Edmonton, Canada
| | - Yonglie Ma
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Helen Nichol
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
84
|
Wang J, Wang G, Yi J, Xu Y, Duan S, Li T, Sun XG, Dong L. The effect of monascin on hematoma clearance and edema after intracerebral hemorrhage in rats. Brain Res Bull 2017; 134:24-29. [PMID: 28655601 DOI: 10.1016/j.brainresbull.2017.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/24/2017] [Accepted: 06/21/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE Intracerebral hemorrhage (ICH) is a particularly devastating form of stroke with high mortality and morbidity. Hematomas are the primary cause of neurologic deficits associated with ICH. The products of hematoma are recognized as neurotoxins and the main contributors to edema formation and tissue damage after ICH. Finding a means to efficiently promote absorption of hematoma is a novel clinical challenge for ICH. Peroxisome proliferator-activated receptor gamma (PPARγ) and nuclear factor erythroid 2-related factor 2 (Nrf2), had been shown that, can take potential roles in the endogenous hematoma clearance. However, monascin, a novel natural Nrf2 activator with PPARγ agonist, has not been reported to play a role in ICH. This study was designed to evaluate the effect of monascin on neurological deficits, hematoma clearance and edema extinction in a model of ICH in rats. METHODS 164 adult male Sprague-Dawley (SD) rats were randomly divided into sham; vehicle; monascin groups with low dosages (1mg/kg/day), middle dosages (5mg/kg/day) and high dosages (10mg/kg/day) respectively. Animals were euthanized at 1, 3 and 7days following neurological evaluation after surgery. We examined the effect of monascin on the brain water contents, blood brain barrier (BBB) permeability and hemoglobin levels, meanwhile reassessed the volume of hematoma and edema around the hematoma by Magnetic Resonance Imaging (MRI) in each group. RESULTS The high dosage of monascin significantly improved neurological deficits, reduced the volume of hematoma in 1-7days after ICH, decreased BBB permeability and edema formation in 1-3days following ICH. CONCLUSION Our study demonstrated that the high dosage of monascin played a neuroprotective role in ICH through reducing BBB permeability, edema and hematoma volume.
Collapse
Affiliation(s)
- Juan Wang
- Department of Neurology, Shanxi Medical University, 56 Xinjian S Rd, Yingze, Taiyuan, Shanxi, 030001, China
| | - Gaiqing Wang
- Department of Neurology, Shanxi Medical University, 56 Xinjian S Rd, Yingze, Taiyuan, Shanxi, 030001, China; Department of Neurology, The Second Hospital, Shanxi Medical University, 382 WuYi Avenue, Taiyuan, Shanxi, 030001, China.
| | - Jinying Yi
- Department of Neurology, Shanxi Medical University, 56 Xinjian S Rd, Yingze, Taiyuan, Shanxi, 030001, China
| | - Yi Xu
- Department of Radiology, The Second Hospital, Shanxi Medical University, 382 WuYi Avenue, Taiyuan, Shanxi, 030001, China
| | - Shuna Duan
- Department of Neurology, Shanxi Medical University, 56 Xinjian S Rd, Yingze, Taiyuan, Shanxi, 030001, China
| | - Tong Li
- Department of Neurology, Shanxi Medical University, 56 Xinjian S Rd, Yingze, Taiyuan, Shanxi, 030001, China
| | - Xin-Gang Sun
- Department of Neurology, The Second Hospital, Shanxi Medical University, 382 WuYi Avenue, Taiyuan, Shanxi, 030001, China
| | - Liang Dong
- Department of Neurology, Shanxi Medical University, 56 Xinjian S Rd, Yingze, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
85
|
Rosiglitazone pretreatment influences thrombin-induced phagocytosis by rat microglia via activating PPARγ and CD36. Neurosci Lett 2017; 651:159-164. [DOI: 10.1016/j.neulet.2017.04.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/17/2017] [Accepted: 04/20/2017] [Indexed: 11/18/2022]
|
86
|
Duan XC, Wang W, Feng DX, Yin J, Zuo G, Chen DD, Chen ZQ, Li HY, Wang Z, Chen G. Roles of autophagy and endoplasmic reticulum stress in intracerebral hemorrhage-induced secondary brain injury in rats. CNS Neurosci Ther 2017; 23:554-566. [PMID: 28544790 DOI: 10.1111/cns.12703] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/23/2017] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES This study aimed to evaluate the roles of autophagy and endoplasmic reticulum (ER) stress in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI) in rats. METHODS Autophagy inducer (rapamycin) and inhibitor (3-methyladenine), as well as ER stress activator (tunicamycin, TM) and inhibitor (tauroursodeoxycholic acid, TUDCA), were used. Bafilomycin A1, an inhibitor of autophagosome-lysosome fusion, was used to assess autophagic flux. RESULTS Autophagy and ER stress were enhanced in the week after ICH. At 6 hours after ICH, autophagy was excessive, while the autophagic flux was damaged at 72 hours and return to be intact at 7 days after ICH. At 6 hours after ICH, ER stress induction by TM could enhance autophagy and lead to caspase 12-mediated apoptosis and neuronal degeneration, which was further aggravated by autophagy induction. At 7 days after ICH, ER stress inhibition by TUDCA still could suppress ICH-induced SBI. And, the effects of TUDCA were enhanced by autophagy induction. CONCLUSIONS At 6 hours after ICH, excessive autophagy may participate in ER stress-induced brain injury; at 7 days after ICH, autophagy could enhance the protection of ER stress inhibitor possibly via clearing up the cell rubbish generated due to the early-stage damaged autophagic flux.
Collapse
Affiliation(s)
- Xiao-Chun Duan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Jia Yin
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Zuo
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dong-Dong Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhou-Qing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Ying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
87
|
Signal transduction involved in lipoxin A4‑induced protection of tubular epithelial cells against hypoxia/reoxygenation injury. Mol Med Rep 2017; 15:1682-1692. [PMID: 28259922 PMCID: PMC5365021 DOI: 10.3892/mmr.2017.6195] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 12/15/2016] [Indexed: 11/05/2022] Open
Abstract
Previous studies have reported that lipoxin A4 (LXA4) may exert a renoprotective effect on ischemia/reperfusion injury in various animal models. The underlying mechanism of LXA4-induced renoprotection during ischemia/reperfusion injury remains to be elucidated. The present study investigated LXA4-induced protection on renal tubular cells subjected to hypoxia/reoxygenation (H/R) injury, and determined the effects of peroxisome proliferator-activated receptor-γ (PPARγ) and heme oxygenase-1 (HO-1) on LXA4 treatment. HK-2 human tubular epithelial cells exposed to H/R injury were pretreated with LXA4, signal molecule inhibitors or the HO-1 inhibitor zinc protoporphyrin-IX, or were transfected with PPARγ small interfering RNA (siRNA) or nuclear factor E2-related factor 2 (Nrf2) siRNA. The protein and mRNA expression levels of PPARγ and HO-1 were analyzed using western blotting and reverse transcription-quantitative polymerase chain reaction. Binding activity of Nrf2 to the HO-1 E1 enhancer was determined using chromatin immunoprecipitation. Nrf2 binding to the HO-1 antioxidant responsive element (ARE) was assessed using electrophoretic mobility shift assay. Preincubation of cells with LXA4 exposed to H/R injury led to a decreased production of inducible nitrogen oxide synthase, malondialdehyde, γ-glutamyl transpeptidase, leucine aminopeptidase and N-acetyl-β-glucosaminidase. In addition, LXA4 pretreatment increased cell viability, protein and mRNA expression levels of PPARγ and HO-1 and PPARγ and HO-1 promoter activity. SB20358 is a p38 mitogen-activated protein kinase (p38 MAPK) pathway inhibitor, which reduced LXA4-induced PPARγ expression levels. LXA4 treatment upregulated p38 MAPK activation, Nrf2 nuclear translocation and increased binding activity of Nrf2 to HO-1 ARE and E1 enhancer in cells exposed to H/R injury. Transfection of the cells with PPARγ siRNA reduced the LXA4-induced Nrf2 translocation. Transfection of the cells with PPARγ siRNA or Nrf2 siRNA also reduced the LXA4-induced increase in HO-1 expression. In conclusion, LXA4-induced protection of renal tubular cells against H/R injury was associated with the induction of PPARγ and HO-1, via activation of the p38 MAPK pathway, as well as Nrf2 nuclear translocation and binding to HO-1 ARE and E1 enhancer. Therefore, LXA4-induced renoprotection is associated with activation of the p38 MAPK/PPARγ/Nrf2-ARE/HO-1 pathway.
Collapse
|
88
|
Lu K, Liu G, Yang L, Liu F, Gao L, Shi J, Deng X, Li Q, Xu D, Shi S. Sustainable inflammation transforms hepatic cells by causing oxidative stress injury and potential epithelial-mesenchymal transition. Int J Oncol 2016; 49:971-80. [PMID: 27315196 DOI: 10.3892/ijo.2016.3580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 05/27/2016] [Indexed: 11/06/2022] Open
Abstract
The inflammatory microenvironment promotes tumorigenesis. However, the mechanism through which inflammation transforms hepatic cells in precancerous lesions remains unclear. Hepatic cells undergo significant changes in metabolism before carcinogenesis, but the specific alterations in gene expression and cellular functions in response to precancerous inflammation have not been elucidated. In this study, a hepatitis-hepatoma mouse model was successfully established. Label-free quantitative (LFQ) proteomics coupled with bioinformatics analysis was then performed to identify differentially expressed proteins and their functions in hepatic cells with precancerous inflammation. We found that different chemical treatments induced several common changes in the model. Hepatic cells underwent serious oxidative stress injury. Canonical pathway analysis using IPA revealed the activation of signaling pathways, such as integrin signaling, signaling by Rho family GTPases, IL-8 signaling, and ILK signaling, as well as the inhibition of RhoGDI signaling. Analysis of the KEGG pathway indicated alteration in the pathways for focal adhesion and regulation of actin cytoskeleton. Results from western blot analysis demonstrated the upregulation of proteins, including p-STAT3, TWIST, SNAIL, Vimentin, and MMP-9, which are involved in epithelial-mesenchymal transition (EMT). These results indicated that hepatic cells were likely to undergo EMT. Interestingly, the expression of E-cadherin was upregulated, but this observation must be further investigated. In conclusion, the results revealed that notable functional and pathway changes occurred during the precancerous inflammation stage in the liver. Our study contributes to understanding of the roles of inflammation in tumorigenesis and provides a molecular basis for further studies on the tumorigenesis of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Kun Lu
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Guoyan Liu
- Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Ling Yang
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Fan Liu
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Libin Gao
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Jingxian Shi
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Xiaoling Deng
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Qifu Li
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Donghui Xu
- Department of Hepatic Biliary Pancreatic Vascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Songlin Shi
- Department of Basic Medicine, Medical College of Xiamen University/Cancer Research Center of Xiamen University, Xiamen, Fujian 361102, P.R. China
| |
Collapse
|
89
|
Taurine protects against As2O3-induced autophagy in livers of rat offsprings through PPARγ pathway. Sci Rep 2016; 6:27733. [PMID: 27291853 PMCID: PMC4904213 DOI: 10.1038/srep27733] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/24/2016] [Indexed: 11/08/2022] Open
Abstract
Chronic exposures to arsenic had been associated with metabolism diseases. Peroxisome proliferator-activated receptor gamma (PPARγ) was found in the liver, regulated metabolism. Here, we found that the expression of PPARγ was decreased, the generation of reactive oxygen species (ROS) and autophagy were increased after treatment with As2O3 in offsprings’ livers. Taurine (Tau), a sulfur-containing β–amino acid could reverse As2O3-inhibited PPARγ. Tau also inhibit the generation of ROS and autophagy. We also found that As2O3 caused autophagic cell death and ROS accelerated in HepG2 cells. Before incubation with As2O3, the cells were pretreated with PPARγ activator Rosiglitazone (RGS), we found that autophagy and ROS was inhibited in HepG2 cells, suggesting that inhibition of PPARγ contributed to As2O3-induced autophagy and the generation of ROS. After pretreatment with Tau, the level of PPARγ was improved and the autophagy and ROS was inhibited in As2O3-treated cells, suggesting that Tau could protect hepatocytes against As2O3 through modulating PPARγ pathway.
Collapse
|
90
|
The Injury and Therapy of Reactive Oxygen Species in Intracerebral Hemorrhage Looking at Mitochondria. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2592935. [PMID: 27293511 PMCID: PMC4880716 DOI: 10.1155/2016/2592935] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/20/2016] [Accepted: 04/17/2016] [Indexed: 01/12/2023]
Abstract
Intracerebral hemorrhage is an emerging major health problem often resulting in death or disability. Reactive oxygen species (ROS) have been identified as one of the major damaging factors in ischemic stroke. However, there is less discussion about ROS in hemorrhage stroke. Metabolic products of hemoglobin, excitatory amino acids, and inflammatory cells are all sources of ROS, and ROS harm the central nervous system through cell death and structural damage, especially disruption of the blood-brain barrier. We have considered the antioxidant system of the CNS itself and the drugs aiming to decrease ROS after ICH, and we find that mitochondria are key players in all of these aspects. Moreover, when the mitochondrial permeability transition pore opens, ROS-induced ROS release, which leads to extensive liberation of ROS and mitochondrial failure, occurs. Therefore, the mitochondrion may be a significant target for elucidating the problem of ROS in ICH; however, additional experimental support is required.
Collapse
|
91
|
Wu G, Wu J, Wang L, Jiao Y, Zhou H, Tang Z. Minimally invasive surgery for ICH evacuation followed by rosiglitazone infusion therapy increased perihematomal PPARγ expression and improved neurological outcomes in rabbits. Neurol Res 2016; 38:261-8. [PMID: 27082035 DOI: 10.1080/01616412.2015.1105627] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To observe the effects of minimally invasive surgery (MIS) for intracerebral hematoma (ICH) evacuation followed by rosiglitazone infusion therapy on peroxisome proliferator-activated receptor-gamma (PPARγ), blood-brain barrier (BBB) permeability, and neurological function. METHODS A total of 75 male rabbits (2.8-3.4 kg) were randomly assigned to a normal control group (NC group), a model control group (MC group), a rosiglitazone group (RSG group), a minimally invasive treatment group (MIS group) or a MIS combined with rosiglitazone group (MIS+RSG group). ICH was induced in all of the animals except for those in the NC group. The rosiglitazone was infused into the hematoma area in the RSG group and the MIS+RSG group. A MIS was performed to evacuate the ICH 6 h after the successful preparation of the ICH model in the MIS group and the MIS+RSG group. Each group included 15 rabbits and was divided equally into 3 subgroups (each subgroup included 5 rabbits that were killed on day 1, day 3, or day 7). Neurological deficit scores were determined, and the perihematomal brain tissue was removed to determine the PPARγ level and BBB permeability. RESULTS Neurological deficit scores, perihematomal PPARγ levels, and BBB permeability were all significantly increased in the MC group compared to the NC group. Performing the MIS alone to evacuate the ICH resulted in a marked decrease in these indices. The RSG used alone increased PPARγ levels and decreased BBB disruption. The MIS+RSG group displayed a marked increase in PPARγ levels and a more significant decrease in BBB permeability and neurological deficit scores. CONCLUSIONS Performing MIS followed by PPARγ agonist infusion therapy is more efficacious for reducing secondary damage to the brain and improving neurological function.
Collapse
Affiliation(s)
- Guofeng Wu
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Junjie Wu
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Likun Wang
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Yu Jiao
- a Department of Emergency , Affiliated Hospital of Guizhou Medical University , Guiyang City , Guizhou Province , China
| | - Houguang Zhou
- b Department of Neurology , Affiliated Huashan Hospital of Fudan University , Shanghai , China
| | - Zhouping Tang
- c Department of Neurology , Affiliated Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan City , Hubei Province , China
| |
Collapse
|
92
|
Intracerebral Hemorrhage, Oxidative Stress, and Antioxidant Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1203285. [PMID: 27190572 PMCID: PMC4848452 DOI: 10.1155/2016/1203285] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/20/2015] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
Hemorrhagic stroke is a common and severe neurological disorder and is associated with high rates of mortality and morbidity, especially for intracerebral hemorrhage (ICH). Increasing evidence demonstrates that oxidative stress responses participate in the pathophysiological processes of secondary brain injury (SBI) following ICH. The mechanisms involved in interoperable systems include endoplasmic reticulum (ER) stress, neuronal apoptosis and necrosis, inflammation, and autophagy. In this review, we summarized some promising advances in the field of oxidative stress and ICH, including contained animal and human investigations. We also discussed the role of oxidative stress, systemic oxidative stress responses, and some research of potential therapeutic options aimed at reducing oxidative stress to protect the neuronal function after ICH, focusing on the challenges of translation between preclinical and clinical studies, and potential post-ICH antioxidative therapeutic approaches.
Collapse
|
93
|
Katsuyama Y, Tsuboi T, Taira N, Yoshioka M, Masaki H. 3-O-Laurylglyceryl ascorbate activates the intracellular antioxidant system through the contribution of PPAR-γ and Nrf2. J Dermatol Sci 2016; 82:189-96. [PMID: 26976686 DOI: 10.1016/j.jdermsci.2016.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 02/25/2016] [Accepted: 03/02/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Ascorbic acid (AsA) has multifunctional effects on physiology and aging including the prevention and improvement of skin pigmentation and wrinkles. AsA has scavenging effects against various types of reactive oxygen species (ROS), which are initiators of aging and premature aging of the skin. However, AsA not only has a quite unstable characteristic, but also has low skin penetration. In addition, existing water-soluble AsA derivatives are not effective to improve its penetration of the skin. OBJECTIVE To investigate the antioxidant effect of a newly synthesized amphipathic derivative of AsA, 3-O-laurylglyceryl ascorbate (VC-3LG), in which a laurylglyceryl group was introduced into AsA. METHODS Intracellular ROS levels in keratinocytes were evaluated using the 2',7'-Dichlorofluorescein diacetate (DCFHDA) assay. Real-time PCR was used to investigate the mechanism of the antioxidant effect of VC-3LG. RESULTS Although VC-3LG had less ability to scavenge ROS compared to AsA, it elicited a superior reduction of intracellular ROS levels, with or without extracellular stimuli such as exposure to H2O2 or UVB. The results show that VC-3LG up-regulates the expression of mRNAs encoding peroxisome proliferator activated receptor-γ (PPAR-γ) and nuclear factor E2-related factor 2 (Nrf2), which in turn up-regulate the levels of mRNAs encoding γ-glutamyl cysteine synthase (γ-GCS), heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase-1 (NQO1). Furthermore, the Nrf2 mRNA level is down-regulated in siPPAR-γ treated cells, and the effects of VC-3LG on PPAR-γ and Nrf2 mRNA levels are reduced by PPAR-γ knockdown. CONCLUSION Taken together, we conclude that VC-3LG has an antioxidant effect and scavenges ROS directly as well as stimulating intracellular antioxidants such as GSH through the PPAR-γ and Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yushi Katsuyama
- Seiwa Kasei Co., Ltd., 1-2-14, Nunoichicho, Higashiosaka, Osaka 579-8004, Japan.
| | - Tatsuya Tsuboi
- Seiwa Kasei Co., Ltd., 1-2-14, Nunoichicho, Higashiosaka, Osaka 579-8004, Japan
| | - Norihisa Taira
- Seiwa Kasei Co., Ltd., 1-2-14, Nunoichicho, Higashiosaka, Osaka 579-8004, Japan
| | - Masato Yoshioka
- Seiwa Kasei Co., Ltd., 1-2-14, Nunoichicho, Higashiosaka, Osaka 579-8004, Japan
| | - Hitoshi Masaki
- Tokyo University of Technology, 1404-1, Katakuracho, Hachiouji, Tokyo 192-0982, Japan
| |
Collapse
|
94
|
Oxidative Stress in Intracerebral Hemorrhage: Sources, Mechanisms, and Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3215391. [PMID: 26843907 PMCID: PMC4710930 DOI: 10.1155/2016/3215391] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is associated with the highest mortality and morbidity despite only constituting approximately 10–15% of all strokes. Complex underlying mechanisms consisting of cytotoxic, excitotoxic, and inflammatory effects of intraparenchymal blood are responsible for its highly damaging effects. Oxidative stress (OS) also plays an important role in brain injury after ICH but attracts less attention than other factors. Increasing evidence has demonstrated that the metabolite axis of hemoglobin-heme-iron is the key contributor to oxidative brain damage after ICH, although other factors, such as neuroinflammation and prooxidases, are involved. This review will discuss the sources, possible molecular mechanisms, and potential therapeutic targets of OS in ICH.
Collapse
|
95
|
Gliem M, Klotz L, van Rooijen N, Hartung HP, Jander S. Hyperglycemia and PPARγ Antagonistically Influence Macrophage Polarization and Infarct Healing After Ischemic Stroke. Stroke 2015; 46:2935-42. [PMID: 26337971 DOI: 10.1161/strokeaha.115.010557] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/11/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Secondary intracerebral hemorrhage (sICH) is a potentially serious complication of ischemic stroke, in particular under concomitant oral anticoagulation. Previous studies in murine stroke models defined a novel vascular repair function of hematogenous monocytes/macrophages (MO/MP), which proved essential for the prevention of oral anticoagulation-associated sICH. Here, we addressed the question whether hyperglycemia as a clinically relevant prohemorrhagic risk factor and peroxisome proliferator-activated receptor gamma (PPARγ) activation affect MO/MP differentiation and the risk of sICH after ischemic stroke. METHODS Oral anticoagulation-associated sICH was induced by phenprocoumon feeding to mice undergoing transient middle cerebral artery occlusion. Hyperglycemia was induced by streptozotocin treatment. The role of PPARγ-dependent MO/MP differentiation was addressed in mice with myeloid cell-specific PPARγ-knockout (LysM-PPARγ(KO)). Pharmacological PPARγ activation via pioglitazone was tested as a treatment option. RESULTS Hyperglycemic mice and normoglycemic LysM-PPARγ(KO) mice exhibited abnormal proinflammatory skewing of their hematogenous MO/MP response and abnormal vascular remodeling in the infarct border zone, leading to an increased rate of oral anticoagulation-associated sICH. Pharmacological PPARγ activation in hyperglycemic mice corrected the inflammatory response toward an anti-inflammatory profile, stabilized neovessels in the infarct border zone, and reduced the rate of sICH. This preventive effect was dependent on the presence of macrophages, but independent from effects on blood glucose levels. CONCLUSIONS Hyperglycemia and macrophage-specific PPARγ activation exert opposing effects on MO/MP polarization in ischemic stroke lesions and, thereby, critically determine the risk of hemorrhagic infarct transformation.
Collapse
Affiliation(s)
- Michael Gliem
- From the Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany (M.G., H.-P.H., S.J.); Department of Neurology, University of Münster, Münster, Germany (L.K.); and Department of Cell Biology and Immunology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands (N.v.R.)
| | - Luisa Klotz
- From the Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany (M.G., H.-P.H., S.J.); Department of Neurology, University of Münster, Münster, Germany (L.K.); and Department of Cell Biology and Immunology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands (N.v.R.)
| | - Nico van Rooijen
- From the Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany (M.G., H.-P.H., S.J.); Department of Neurology, University of Münster, Münster, Germany (L.K.); and Department of Cell Biology and Immunology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands (N.v.R.)
| | - Hans-Peter Hartung
- From the Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany (M.G., H.-P.H., S.J.); Department of Neurology, University of Münster, Münster, Germany (L.K.); and Department of Cell Biology and Immunology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands (N.v.R.)
| | - Sebastian Jander
- From the Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany (M.G., H.-P.H., S.J.); Department of Neurology, University of Münster, Münster, Germany (L.K.); and Department of Cell Biology and Immunology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands (N.v.R.).
| |
Collapse
|
96
|
Dharap A, Pokrzywa C, Murali S, Kaimal B, Vemuganti R. Mutual induction of transcription factor PPARγ and microRNAs miR-145 and miR-329. J Neurochem 2015; 135:139-46. [PMID: 26119485 DOI: 10.1111/jnc.13220] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/12/2015] [Accepted: 06/17/2015] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are known to control mRNA translation. Most miRNAs are transcribed from specific genes with well-defined promoters located throughout the genome. The mechanisms that control miRNA expression under normal and pathological conditions are not yet understood clearly. Peroxisome proliferator-activated receptor (PPAR) γ is a ligand-activated transcription factor that is extensively distributed in the CNS. PPARγ activation induces neuroprotection by modulating genes that contain peroxisome proliferator response elements (PPREs) in their promoters. We presently evaluated if PPARγ modulates miRNA expression. When adult rats were treated with PPARγ agonist rosiglitazone, expression of 28 miRNAs altered significantly (12 up- and 16 down-regulated; 3-119 fold) in the cerebral cortex compared to vehicle-treated controls. In silico analysis showed 1-5 PPREs in the putative promoter regions (within 1 Kb upstream of the transcription start site) of these miRNA genes. Cotransfection with a PPARγ constitutively expressing vector significantly induced the miR-145 and miR-329 promoter vectors (each have four PPREs), which was curtailed by point mutations of PPREs in their promoters. Interestingly, the PPARγ promoter has binding sites for both these miRNAs and transfection with miR-329 mimic and miR-145 mimic induced the PPARγ expression. Thus, these studies show a cyclical induction of miRNAs and PPARγ, indicating that the pleiotropic beneficial effects of PPARγ agonists might be modulated in part by miRNAs and their down-stream mRNAs. We proposed that promoters of many microRNAs contain the binding sites for the transcription factor PPARγ. Activation of PPARγ modulates the expression of these microRNAs. Two such PPARγ-responsive microRNAs (miR-145 and miR-329) bind to PPARγ promoter to induce its expression. This indicates the presence of a feedback loop by which transcription factors and microRNAs can modulate each other.
Collapse
Affiliation(s)
- Ashutosh Dharap
- Department of Neurological Surgery and Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Courtney Pokrzywa
- Department of Neurological Surgery and Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Shruthi Murali
- Department of Neurological Surgery and Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Balarama Kaimal
- Department of Neurological Surgery and Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery and Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
97
|
Mahmoud-Awny M, Attia AS, Abd-Ellah MF, El-Abhar HS. Mangiferin Mitigates Gastric Ulcer in Ischemia/ Reperfused Rats: Involvement of PPAR-γ, NF-κB and Nrf2/HO-1 Signaling Pathways. PLoS One 2015. [PMID: 26196679 PMCID: PMC4509761 DOI: 10.1371/journal.pone.0132497] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mangiferin (MF), a xanthonoid from Mangifera indica, has been proved to have antisecretory and antioxidant gastroprotective effects against different gastric ulcer models; however, its molecular mechanism has not been previously elucidated. Therefore, the aim of this study was to test its modulatory effect on several signaling pathways using the ischemia/reperfusion model for the first time. Animals were treated with MF, omeprazole (OMP), and the vehicle. The mechanistic studies revealed that MF mediated its gastroprotective effect partly via inducing the expression of Nrf2, HO-1 and PPAR-γ along with downregulating that of NF-κB. Surprisingly, the effect of MF, especially the high dose, exceeded that mediated by OMP except for Nrf2. The molecular results were reflected on the biomarkers measured, where the antioxidant effect of MF was manifested by increasing total antioxidant capacity and glutathione, besides normalizing malondialdehyde level. Additionally, MF decreased the I/R-induced nitric oxide elevation, an effect that was better than that of OMP. In the serum, MF, dose dependently, enhanced endothelial nitric oxide synthase, while reduced the inducible isoform. Regarding the anti-inflammatory effect of MF, it reduced serum level of IL-1β and sE-selectin, effects that were mirrored on the tissue level of myeloperoxidase, the neutrophil infiltration marker. In addition, MF possessed an antiapoptotic character evidenced by elevating Bcl-2 level and reducing that of caspase-3 in a dose related order. As a conclusion, the intimated gastroprotective mechanisms of MF are mediated, partially, by modulation of oxidative stress, inflammation and apoptosis possibly via the Nrf2/HO-1, PPAR-γ/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Magdy Mahmoud-Awny
- Department of Pharmacology & Toxicology, October University, Cairo, Egypt
| | - Ahmed S. Attia
- Department of Microbiology & Immunologyology, Cairo University, Cairo, Egypt
| | | | - Hanan Salah El-Abhar
- Department of Pharmacology & Toxicology, Cairo University, Cairo, Egypt
- * E-mail:
| |
Collapse
|
98
|
Leviton A, Gressens P, Wolkenhauer O, Dammann O. Systems approach to the study of brain damage in the very preterm newborn. Front Syst Neurosci 2015; 9:58. [PMID: 25926780 PMCID: PMC4396381 DOI: 10.3389/fnsys.2015.00058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/26/2015] [Indexed: 12/11/2022] Open
Abstract
Background: A systems approach to the study of brain damage in very preterm newborns has been lacking. Methods: In this perspective piece, we offer encephalopathy of prematurity as an example of the complexity and interrelatedness of brain-damaging molecular processes that can be initiated inflammatory phenomena. Results: Using three transcription factors, nuclear factor-kappa B (NF-κB), Notch-1, and nuclear factor erythroid 2 related factor 2 (NRF2), we show the inter-connectedness of signaling pathways activated by some antecedents of encephalopathy of prematurity. Conclusions: We hope that as biomarkers of exposures and processes leading to brain damage in the most immature newborns become more readily available, those who apply a systems approach to the study of neuroscience can be persuaded to study the pathogenesis of brain disorders in the very preterm newborn.
Collapse
Affiliation(s)
- Alan Leviton
- Neuroepidemiology Unit, Boston Children's Hospital Boston, MA, USA ; Department of Neurology, Harvard Medical School Boston, MA, USA
| | - Pierre Gressens
- Inserm, U1141 Paris, France ; Department of Perinatal Imaging and Health, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital London, UK
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock Rostock, Germany ; Stellenbosch Institute for Advanced Study (STIAS) Stellenbosch, South Africa
| | - Olaf Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine Boston, MA, USA ; Perinatal Epidemiology Unit, Department of Gynecology and Obstetrics, Hannover Medical School Hannover, Germany
| |
Collapse
|