1
|
Xie S, Li F. Ependymal cells: roles in central nervous system infections and therapeutic application. J Neuroinflammation 2024; 21:255. [PMID: 39385253 PMCID: PMC11465851 DOI: 10.1186/s12974-024-03240-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.
Collapse
Affiliation(s)
- Shiqi Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, 130 Dong An Road, Xuhui District, Shanghai, China.
- Tuberculosis Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
2
|
Tastan B, Heneka MT. The impact of neuroinflammation on neuronal integrity. Immunol Rev 2024; 327:8-32. [PMID: 39470038 DOI: 10.1111/imr.13419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Neuroinflammation, characterized by a complex interplay among innate and adaptive immune responses within the central nervous system (CNS), is crucial in responding to infections, injuries, and disease pathologies. However, the dysregulation of the neuroinflammatory response could significantly affect neurons in terms of function and structure, leading to profound health implications. Although tremendous progress has been made in understanding the relationship between neuroinflammatory processes and alterations in neuronal integrity, the specific implications concerning both structure and function have not been extensively covered, with the exception of perspectives on glial activation and neurodegeneration. Thus, this review aims to provide a comprehensive overview of the multifaceted interactions among neurons and key inflammatory players, exploring mechanisms through which inflammation influences neuronal functionality and structural integrity in the CNS. Further, it will discuss how these inflammatory mechanisms lead to impairment in neuronal functions and architecture and highlight the consequences caused by dysregulated neuronal functions, such as cognitive dysfunction and mood disorders. By integrating insights from recent research findings, this review will enhance our understanding of the neuroinflammatory landscape and set the stage for future interventions that could transform current approaches to preserve neuronal integrity and function in CNS-related inflammatory conditions.
Collapse
Affiliation(s)
- Bora Tastan
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| |
Collapse
|
3
|
Zayeri ZD, Torabizadeh M, Kargar M, Kazemi H. The molecular fingerprint of neuroinflammation in COVID-19: A comprehensive discussion on molecular mechanisms of neuroinflammation due to SARS-COV2 antigens. Behav Brain Res 2024; 462:114868. [PMID: 38246395 DOI: 10.1016/j.bbr.2024.114868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/13/2024] [Accepted: 01/14/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Severe acute respiratory syndrome coronavirus 2 attacks the neural system directly and indirectly via various systems, such as the nasal cavity, olfactory system, and facial nerves. Considering the high energy requirement, lack of antioxidant defenses, and high amounts of metal ions in the brain, oxidative damage is very harmful to the brain. Various neuropathic pain conditions, neurological disorders, and neuropsychiatric complications were reported in Coronavirus disease 2019, prolonged Coronavirus disease 2019, and after Coronavirus disease 2019 immunization. This manuscript offers a distinctive outlook on the interconnectedness between neurology and neuropsychiatry through its meticulous analysis of complications. DISCUSSION After recovering from Coronavirus disease 2019, approximately half of the patients reported developing Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Long Coronavirus disease 2019 imaging reports illustrated the hypometabolism in various parts of the brain, such as olfactory bulbs, limbic/paralimbic domains, the brainstem, and the cerebellum. Ninety imaging and neuropathological studies of Coronavirus disease 2019 have shown evidence of white matter, brainstem, frontotemporal, and oculofrontal lesions. Emotional functions, such as pleasant, long/short-term memory, movement, cognition and cognition in decision-making are controlled by these regions. The neuroinflammation and the mechanisms of defense are well presented in the discussion. The role of microglia activation, Inducible NO synthase, Cyclooxygenases ½, Reactive oxygen species, neurotoxic toxins and pro-inflammatory cytokines, such as Interleukin-1 beta, Interleukin-6 and Tumor Necrosis Factor-alpha are highlighted in neuronal dysfunction and death. Nuclear factor kappa-light-chain-enhancer of activated B cells, Mitogen-activated protein kinase, Activator Protein 1, and Interferon regulatory factors are the main pathways involved in microglia activation in Coronavirus disease 2019 neuroinflammation. CONCLUSION The neurological aspect of Coronavirus disease 2019 should be highlighted. Neurological, psychological, and behavioral aspects of Coronavirus disease 2019, prolonged Coronavirus disease 2019, and Coronavirus disease 2019 vaccines can be the upcoming issues. We need a global awareness where this aspect of the disease should be more considered in health research.
Collapse
Affiliation(s)
- Zeinab Deris Zayeri
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mehdi Torabizadeh
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Kargar
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hashem Kazemi
- Department of Biology, Dezful Branch, Islamic Azad University, Dezful, Iran
| |
Collapse
|
4
|
Zhou X, Wu X, Wang R, Han L, Li H, Zhao W. Mechanisms of 3-Hydroxyl 3-Methylglutaryl CoA Reductase in Alzheimer's Disease. Int J Mol Sci 2023; 25:170. [PMID: 38203341 PMCID: PMC10778631 DOI: 10.3390/ijms25010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide and has a high incidence in the elderly. Unfortunately, there is no effective therapy for AD owing to its complicated pathogenesis. However, the development of lipid-lowering anti-inflammatory drugs has heralded a new era in the treatment of Alzheimer's disease. Several studies in recent years have shown that lipid metabolic dysregulation and neuroinflammation are associated with the pathogenesis of AD. 3-Hydroxyl 3-methylglutaryl CoA reductase (HMGCR) is a rate-limiting enzyme in cholesterol synthesis that plays a key role in cholesterol metabolism. HMGCR inhibitors, known as statins, have changed from being solely lipid-lowering agents to neuroprotective compounds because of their effects on lipid levels and inflammation. In this review, we first summarize the main regulatory mechanism of HMGCR affecting cholesterol biosynthesis. We also discuss the pathogenesis of AD induced by HMGCR, including disordered lipid metabolism, oxidative stress, inflammation, microglial proliferation, and amyloid-β (Aβ) deposition. Subsequently, we explain the possibility of HMGCR as a potential target for AD treatment. Statins-based AD treatment is an ascent field and currently quite controversial; therefore, we also elaborate on the current application prospects and limitations of statins in AD treatment.
Collapse
Affiliation(s)
- Xun Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Xiaolang Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Rui Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Lu Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Huilin Li
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| |
Collapse
|
5
|
Wartchow KM, Scaini G, Quevedo J. Glial-Neuronal Interaction in Synapses: A Possible Mechanism of the Pathophysiology of Bipolar Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:191-208. [PMID: 36949311 DOI: 10.1007/978-981-19-7376-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Bipolar disorder (BD) is a severe and chronic psychiatric disorder that affects approximately 1-4% of the world population and is characterized by recurrent episodes of mania or hypomania and depression. BD is also associated with illnesses marked by immune activation, such as metabolic syndrome, obesity, type 2 diabetes mellitus, and cardiovascular diseases. Indeed, a connection has been suggested between neuroinflammation and peripheral inflammatory markers in the pathophysiology of BD, which can be associated with the modulation of many dysfunctional processes, including synaptic plasticity, neurotransmission, neurogenesis, neuronal survival, apoptosis, and even cognitive/behavioral functioning. Rising evidence suggests that synaptic dysregulations, especially glutamatergic system dysfunction, are directly involved in mood disorders. It is becoming clear that dysregulations in connection and structural changes of glial cells play a central role in the BD pathophysiology. This book chapter highlighted the latest findings that support the theory of synaptic dysfunction in BD, providing an overview of the alterations in neurotransmitters release, astrocytic uptake, and receptor signaling, as well as the role of inflammation on glial cells in mood disorders. Particular emphasis is given to the alterations in presynaptic and postsynaptic neurons and glial cells, all cellular elements of the "tripartite synapse," compromising the neurotransmitters system, excitatory-inhibitory balance, and neurotrophic states of local networks in mood disorders. Together, these studies provide a foundation of knowledge about the exact role of the glial-neuronal interaction in mood disorders.
Collapse
Affiliation(s)
- Krista M Wartchow
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Giselli Scaini
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - João Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
6
|
Nelles DG, Hazrati LN. Ependymal cells and neurodegenerative disease: outcomes of compromised ependymal barrier function. Brain Commun 2022; 4:fcac288. [PMID: 36415662 PMCID: PMC9677497 DOI: 10.1093/braincomms/fcac288] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/13/2022] [Accepted: 11/01/2022] [Indexed: 08/08/2023] Open
Abstract
Within the central nervous system, ependymal cells form critical components of the blood-cerebrospinal fluid barrier and the cerebrospinal fluid-brain barrier. These barriers provide biochemical, immunological and physical protection against the entry of molecules and foreign substances into the cerebrospinal fluid while also regulating cerebrospinal fluid dynamics, such as the composition, flow and removal of waste from the cerebrospinal fluid. Previous research has demonstrated that several neurodegenerative diseases, such as Alzheimer's disease and multiple sclerosis, display irregularities in ependymal cell function, morphology, gene expression and metabolism. Despite playing key roles in maintaining overall brain health, ependymal barriers are largely overlooked and understudied in the context of disease, thus limiting the development of novel diagnostic and treatment options. Therefore, this review explores the anatomical properties, functions and structures that define ependymal cells in the healthy brain, as well as the ways in which ependymal cell dysregulation manifests across several neurodegenerative diseases. Specifically, we will address potential mechanisms, causes and consequences of ependymal cell dysfunction and describe how compromising the integrity of ependymal barriers may initiate, contribute to, or drive widespread neurodegeneration in the brain.
Collapse
Affiliation(s)
- Diana G Nelles
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, 555 University Ave, Canada
| | - Lili-Naz Hazrati
- Correspondence to: Dr. Lili-Naz Hazrati 555 University Ave, Toronto ON M5G 1X8, Canada E-mail:
| |
Collapse
|
7
|
Mi L, Min X, Chai Y, Zhang J, Chen X. NLRP1 Inflammasomes: A Potential Target for the Treatment of Several Types of Brain Injury. Front Immunol 2022; 13:863774. [PMID: 35707533 PMCID: PMC9189285 DOI: 10.3389/fimmu.2022.863774] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/28/2022] [Indexed: 12/28/2022] Open
Abstract
NOD-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) is a member of the NLR family. The NLRP1 inflammasome consists of the NLRP1 protein, the adaptor protein apoptosis-associated speck-like protein containing a CARD domain, and the effector molecule pro-caspase-1. When stimulated, the inflammasome initiates the cleavage of pro-caspase-1 and converts it into its active form, caspase-1; then, caspase-1 facilitates the cleavage of the proinflammatory cytokines interleukin-1β and interleukin-18 into their active and secreted forms. In addition, caspase-1 also mediates the cleavage of gasdermin D, which leads to pyroptosis, an inflammatory form of cell death. Pathological events that damage the brain and result in neuropathological conditions can generally be described as brain injury. Neuroinflammation, especially that driven by NLRP1, plays a considerable role in the pathophysiology of brain injury, such as early brain injury (EBI) of subarachnoid hemorrhage, ischemic brain injury during stroke, and traumatic brain injury (TBI). In this article, a thorough overview of NLRP1 is presented, including its structure, mechanism of activation, and role in neuroinflammation. We also present recent studies on NLRP1 as a target for the treatment of EBI, ischemic brain injury, TBI, and other types of brain injury, thus highlighting the perspective of NLRP1 as an effective mediator of catastrophic brain injury.
Collapse
Affiliation(s)
- Liang Mi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xiaobin Min
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Baodi Clinical College, Tianjin Medical University, Tianjin, China
| | - Yan Chai
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- *Correspondence: Xin Chen,
| |
Collapse
|
8
|
Katt WP, Aplin C, Cerione RA. Exploring the Role of Transglutaminase in Patients with Glioblastoma: Current Perspectives. Onco Targets Ther 2022; 15:277-290. [PMID: 35340676 PMCID: PMC8943831 DOI: 10.2147/ott.s329262] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 03/08/2022] [Indexed: 12/22/2022] Open
Abstract
Tissue transglutaminase (tTG) is a rather unique GTP-binding/protein crosslinking enzyme that has been shown to play important roles in a number of cellular processes that impact both normal physiology and disease states. This is especially the case in the context of aggressive brain tumors, such as glioblastoma. The diverse roles played by tTG in cancer survival and progression have led to significant interest in recent years in using tTG as a therapeutic target. In this review, we provide a brief overview of the transglutaminase family, and then discuss the primary biochemical activities exhibited by tTG with an emphasis on the role it plays in glioblastoma progression. Finally, we consider current approaches to target tTG which might eventually have clinical impact.
Collapse
Affiliation(s)
- William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Cody Aplin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY, 14850, USA,Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, USA,Correspondence: Richard A Cerione, Tel +1 607-253-3650, Email
| |
Collapse
|
9
|
Roshdy K, Morsy K, Abumandour MMA. Microscopic focus on ependymal cells of the spinal cord of the one-humped camel (Camelus dromedarius): Histological, immunohistochemical, and transmission microscopic study. Microsc Res Tech 2021; 85:1238-1247. [PMID: 34817902 DOI: 10.1002/jemt.23990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/03/2021] [Accepted: 10/17/2021] [Indexed: 11/07/2022]
Abstract
The current study was designed to give a complete microscopic description of the ependymal cells of the one-humped camel (Camelus dromedarius) using histological, immunohistochemical, and transmission microscopic descriptions of the ependymal cells of the fresh 35 spinal cord samples immediately after their slaughtering. In our findings, the central canal of the spinal cord was lined by multilayered stratified cuboidal or columnar ependymal cells. The ependymal cells had an irregular striated border at their free surface. The ependymal cells do not exhibit a basement membrane. The simple oval nucleus was occupied a large part of the cell with spherical mitochondria. The apical surface of the ependymal cells possesses long cilia; each cilium was bounded by an evagination of the luminal plasma membrane. Some ependymal cells had minute finger-like projections on their luminal plasma membrane. In the perinuclear zone of ependymal cells, many cristiform mitochondria, free ribosomes, and Golgi complexes usually occur. Vacuoles with homogenous and clear fluid were observed. The lateral surface of the adjacent ependymal cells exhibits several tight junctions represented by zonulae occludens and adherens. There were many desmosomes between the neighboring ependymal cells. A perinuclear whorl of filaments fills the lateral part of these ependymal cells. The ependymal cells revealed a clear immunohistochemical reaction with proliferating cell nuclear antigen and nestin stain. There were no obvious differences between the different segments of the spinal cord. Our data concluded that the ependymal cells display clear differences in anatomy as well as ultrastructure that may reflect their distinct functional activity.
Collapse
Affiliation(s)
- Karam Roshdy
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Mohamed M A Abumandour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
10
|
Baumgartner JE, Baumgartner LS, Baumgartner ME, Moore EJ, Messina SA, Seidman MD, Shook DR. Progenitor cell therapy for acquired pediatric nervous system injury: Traumatic brain injury and acquired sensorineural hearing loss. Stem Cells Transl Med 2021; 10:164-180. [PMID: 33034162 PMCID: PMC7848325 DOI: 10.1002/sctm.20-0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
While cell therapies hold remarkable promise for replacing injured cells and repairing damaged tissues, cell replacement is not the only means by which these therapies can achieve therapeutic effect. For example, recent publications show that treatment with varieties of adult, multipotent stem cells can improve outcomes in patients with neurological conditions such as traumatic brain injury and hearing loss without directly replacing damaged or lost cells. As the immune system plays a central role in injury response and tissue repair, we here suggest that multipotent stem cell therapies achieve therapeutic effect by altering the immune response to injury, thereby limiting damage due to inflammation and possibly promoting repair. These findings argue for a broader understanding of the mechanisms by which cell therapies can benefit patients.
Collapse
Affiliation(s)
- James E. Baumgartner
- Advent Health for ChildrenOrlandoFloridaUSA
- Department of Neurological SurgeryUniversity of Central Florida College of MedicineOrlandoFloridaUSA
| | | | | | - Ernest J. Moore
- Department of Audiology and Speech Language PathologyUniversity of North TexasDentonTexasUSA
| | | | - Michael D. Seidman
- Advent Health CelebrationCelebrationFloridaUSA
- Department of OtorhinolaryngologyUniversity of Central FloridaOrlandoFloridaUSA
| | | |
Collapse
|
11
|
Kinra M, Joseph A, Nampoothiri M, Arora D, Mudgal J. Inhibition of NLRP3-inflammasome mediated IL-1β release by phenylpropanoic acid derivatives: in-silico and in-vitro approach. Eur J Pharm Sci 2020; 157:105637. [PMID: 33171231 DOI: 10.1016/j.ejps.2020.105637] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022]
Abstract
NLRP3 inflammasome activation and subsequent release of IL-1β are being explored as a causal pathology for inflammatory and autoimmune disorders. Modulation of this pathway by the compounds from natural sources may provide a better targeted approach with improved therapeutic outcome. The study was carried out to test the ability of phenylpropanoic acid derivatives to inhibit the NLRP3 inflammasome pathway and IL-1β release. The main purpose of the study was to test the active derivatives with respect to the possible molecular interactions in-silico, effect on mRNA expression of molecular markers and, effect on released cytokine. Autodock along with SwissADME was used to carry out the in-silico studies including the prediction studies as well as molecular docking studies. The effect of test compounds on mRNA expression of important proteins was evaluated against U87MG cells using RT-qPCR. The changes in released cytokine levels was evaluated using ELISA. The tested phenylpropanoic acid derivatives had a comparable molecular docking profile to that of selected standards. The prediction studies indicated that these compounds have suitable properties to be a drug candidate. mRNA expression studies showed that the derivatives can downregulate the proteins responsible for inflammasome activation and same was reflected in ELISA when the concentration of released cytokine was evaluated. Based on the above results, phenylpropanoic acid derivatives have potential to be developed as specific NLRP3-inflammasome inhibitors.
Collapse
Affiliation(s)
- Manas Kinra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Devinder Arora
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; School of Pharmacy and Pharmacology, MHIQ, QUM Network, Griffith University, Queensland, Australia
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
12
|
Ibrahim AM, Pottoo FH, Dahiya ES, Khan FA, Kumar JBS. Neuron‐glia interactions: Molecular basis of alzheimer’s disease and applications of neuroproteomics. Eur J Neurosci 2020; 52:2931-2943. [DOI: 10.1111/ejn.14838] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Abdallah Mohammad Ibrahim
- Fundamentals of Nursing Department College of Nursing Imam Abdulrahman Bin Faisal University Dammam Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology College of Clinical Pharmacy Imam Abdulrahman Bin Faisal University Dammam Saudi Arabia
| | - Ekta Singh Dahiya
- National Institute of Stroke and Applied Neurosciences (NISAN) Auckland University of Technology Auckland New‐Zealand
| | - Firdos Alam Khan
- Department of Stem Cell Research Institute for Research and Medical Consultations Imam Abdulrahman Bin Faisal University Dammam Saudi Arabia
| | - J. B. Senthil Kumar
- Special centre for Molecular Medicine Jawaharlal Nehru University New‐Delhi India
| |
Collapse
|
13
|
The Role of Neuronal NLRP1 Inflammasome in Alzheimer's Disease: Bringing Neurons into the Neuroinflammation Game. Mol Neurobiol 2019; 56:7741-7753. [PMID: 31111399 DOI: 10.1007/s12035-019-1638-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
The innate immune system and inflammatory response in the brain have critical impacts on the pathogenesis of many neurodegenerative diseases including Alzheimer's disease (AD). In the central nervous system (CNS), the innate immune response is primarily mediated by microglia. However, non-glial cells such as neurons could also partake in inflammatory response independently through inflammasome signalling. The NLR family pyrin domain-containing 1 (NLRP1) inflammasome in the CNS is primarily expressed by pyramidal neurons and oligodendrocytes. NLRP1 is activated in response to amyloid-β (Aβ) aggregates, and its activation subsequently cleaves caspase-1 into its active subunits. The activated caspase-1 proteolytically processes interleukin-1β (IL-1β) and interleukin-18 (IL-18) into maturation whilst co-ordinately triggers caspase-6 which is responsible for apoptosis and axonal degeneration. In addition, caspase-1 activation induces pyroptosis, an inflammatory form of programmed cell death. Studies in murine AD models indicate that the Nlrp1 inflammasome is indeed upregulated in AD and neuronal death is observed leading to cognitive decline. However, the mechanism of NLRP1 inflammasome activation in AD is particularly elusive, given its structural and functional complexities. In this review, we examine the implications of the human NLRP1 inflammasome and its signalling pathways in driving neuroinflammation in AD.
Collapse
|
14
|
Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol 2019; 320:112957. [PMID: 31108085 DOI: 10.1016/j.expneurol.2019.112957] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of both acute and long-term morbidity in the pediatric population, leading to a substantial, long-term socioeconomic burden. Despite the increase in the amount of pre-clinical and clinical research, treatment options for TBI rely heavily on supportive care with very limited targeted interventions that improve the acute and chronic sequelae of TBI. Other than injury prevention, not much can be done to limit the primary injury, which consists of tissue damage and cellular destruction. Secondary injury is the result of the ongoing complex inflammatory pathways that further exacerbate tissue damage, resulting in the devastating chronic outcomes of TBI. On the other hand, some level of inflammation is essential for neuronal regeneration and tissue repair. In this review article we discuss the various stages of the neuroimmune response in the immature, pediatric brain in the context of normal maturation and development of the immune system. The developing brain has unique features that distinguish it from the adult brain, and the immune system plays an integral role in CNS development. Those features could potentially make the developing brain more susceptible to worse outcomes, both acutely and in the long-term. The neuroinflammatory reaction which is triggered by TBI can be described as a highly intricate interaction between the cells of the innate and the adaptive immune systems. The innate immune system is triggered by non-specific danger signals that are released from damaged cells and tissues, which in turn leads to neutrophil infiltration, activation of microglia and astrocytes, complement release, as well as histamine release by mast cells. The adaptive immune response is subsequently activated leading to the more chronic effects of neuroinflammation. We will also discuss current attempts at modulating the TBI-induced neuroinflammatory response. A better understanding of the role of the immune system in normal brain development and how immune function changes with age is crucial for designing therapies to appropriately target the immune responses following TBI in order to enhance repair and plasticity.
Collapse
Affiliation(s)
- Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Young Chun
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America.
| |
Collapse
|
15
|
Labelling of individual ependymal areas in the third and fourth ventricle of the human brain: ependymal tables. Biologia (Bratisl) 2019. [DOI: 10.2478/s11756-019-00192-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Panossian A, Seo EJ, Efferth T. Novel molecular mechanisms for the adaptogenic effects of herbal extracts on isolated brain cells using systems biology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 50:257-284. [PMID: 30466987 DOI: 10.1016/j.phymed.2018.09.204] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/29/2018] [Accepted: 09/17/2018] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Adaptogens are natural compounds or plant extracts that increase adaptability and survival of organisms under stress. Adaptogens stimulate cellular and organismal defense systems by activating intracellular and extracellular signaling pathways and expression of stress-activated proteins and neuropeptides. The effects adaptogens on mediators of adaptive stress response and longevity signaling pathways have been reported, but their stress-protective mechanisms are still not fully understood. AIM OF THE STUDY The aim of this study was to identify key molecular mechanisms of adaptogenic plants traditionally used to treat stress and aging-related disorders, i.e., Rhodiola rosea, Eleutherococcus senticosus, Withania somnifera, Rhaponticum carthamoides, and Bryonia alba. MATERIALS AND METHODS To investigate the underlying molecular mechanisms of adaptogens, we conducted RNA sequencing to profile gene expression alterations in T98G neuroglia cells upon treatment of adaptogens and analyzed the relevance of deregulated genes to adaptive stress-response signaling pathways using in silico pathway analysis software. RESULTS AND DISCUSSION At least 88 of the 3516 genes regulated by adaptogens were closely associated with adaptive stress response and adaptive stress-response signaling pathways (ASRSPs), including neuronal signaling related to corticotropin-releasing hormone, cAMP-mediated, protein kinase A, and CREB; pathways related to signaling involving CXCR4, melatonin, nitric oxide synthase, GP6, Gαs, MAPK, neuroinflammation, neuropathic pain, opioids, renin-angiotensin, AMPK, calcium, and synapses; and pathways associated with dendritic cell maturation and G-coupled protein receptor-mediated nutrient sensing in enteroendocrine cells. All samples tested showed significant effects on the expression of genes encoding neurohormones CRH, GNRH, UCN, G-protein-coupled and other transmembrane receptors TLR9, PRLR, CHRNE, GP1BA, PLXNA4, a ligand-dependent nuclear receptor RORA, transmembrane channels, transcription regulators FOS, FOXO6, SCX, STAT5A, ZFPM2, ZNF396, ZNF467, protein kinases MAPK10, MAPK13, MERTK, FLT1, PRKCH, ROS1, TTN), phosphatases PTPRD, PTPRR, peptidases, metabolic enzymes, a chaperone (HSPA6), and other proteins, all of which modulate numerous life processes, playing key roles in several canonical pathways involved in defense response and regulation of homeostasis in organisms. It is for the first time we report that the molecular mechanism of actions of melatonin and plant adaptogens are alike, all adaptogens tested activated the melatonin signaling pathway by acting through two G-protein-coupled membrane receptors MT1 and MT2 and upregulation of the ligand-specific nuclear receptor RORA, which plays a role in intellectual disability, neurological disorders, retinopathy, hypertension, dyslipidemia, and cancer, which are common in aging. Furthermore, melatonin activated adaptive signaling pathways and upregulated expression of UCN, GNRH1, TLR9, GP1BA, PLXNA4, CHRM4, GPR19, VIPR2, RORA, STAT5A, ZFPM2, ZNF396, FLT1, MAPK10, MERTK, PRKCH, and TTN, which were commonly regulated by all adaptogens tested. We conclude that melatonin is an adaptation hormone playing an important role in regulation of homeostasis. Adaptogens presumably worked as eustressors ("stress-vaccines") to activate the cellular adaptive system by inducing the expression of ASRSPs, which then reciprocally protected cells from damage caused by distress. Functional investigation by interactive pathways analysis demonstrated that adaptogens activated ASRSPs associated with stress-induced and aging-related disorders such as chronic inflammation, cardiovascular health, neurodegenerative cognitive impairment, metabolic disorders, and cancer. CONCLUSION This study has elucidated the genome-wide effects of several adaptogenic herbal extracts in brain cells culture. These data highlight the consistent activation of ASRSPs by adaptogens in T98G neuroglia cells. The extracts affected many genes playing key roles in modulation of adaptive homeostasis, indicating their ability to modify gene expression to prevent stress-induced and aging-related disorders. Overall, this study provides a comprehensive look at the molecular mechanisms by which adaptogens exerts stress-protective effects.
Collapse
Affiliation(s)
| | - Ean-Jeong Seo
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, Mainz 55128, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Staudinger Weg 5, Mainz 55128, Germany.
| |
Collapse
|
17
|
Griffiths MR, Botto M, Morgan BP, Neal JW, Gasque P. CD93 regulates central nervous system inflammation in two mouse models of autoimmune encephalomyelitis. Immunology 2018; 155:346-355. [PMID: 29923617 DOI: 10.1111/imm.12974] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/23/2018] [Accepted: 06/13/2018] [Indexed: 01/10/2023] Open
Abstract
Microglia and non-professional immune cells (endothelial cells, neurons) participate in the recognition and removal of pathogens and tissue debris in the injured central nervous system through major pro-inflammatory processes. However, the mechanisms involved in regulating these responses remain ill-characterized. We herein show that CD93, also known as complement C1qRp/AA4 stem cell marker, has an important role in the regulation of inflammatory processes. The role of CD93 was evaluated in two models of neuroinflammation. We used the MOG-experimental autoimmune encephalomyelitis (EAE) model and the antibody-dependent EAE (ADEAE), which were induced in wild-type and CD93 knockout mice. We found that CD93 was highly expressed by neurons, endothelial cells and microglia (ramified >> amoeboid). Astrocytes and oligodendrocytes did not to express CD93. We further observed that CD93-deficient (CD93-/- ) mice presented a more robust brain and spinal cord inflammation in EAE and ADEAE. Encephalitis in CD93-/- was characterized by increased numbers of infiltrating M1 macrophages (CD11c+ CD206- ) and amoeboid microglia exhibiting a more activated phenotype (Tomato Lectinhigh Cox2high ). Damage to and leakage through the blood-brain barrier was increased in CD93-/- animals and was associated with a more robust neuronal injury when compared with wild-type EAE mice. We propose that CD93 is an important neuro-immune regulator to control central nervous system inflammation.
Collapse
Affiliation(s)
- Mark R Griffiths
- BIIG, Brain Inflammation and Immunity Group, Cardiff University School of Medicine, Cardiff, UK
| | - Marina Botto
- Centre for Complement and Inflammation Research, Department of Medicine, Imperial College, London, UK
| | - Bryan Paul Morgan
- Complement Biology Group, Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - James W Neal
- Neuropathology Department, Cardiff University School of Medicine, Cardiff, UK
| | - Philippe Gasque
- BIIG, Brain Inflammation and Immunity Group, Cardiff University School of Medicine, Cardiff, UK.,GRI EA4517, Immunopathology and infectious disease grouping, CHU, CYROI, Université de La Réunion, Sainte-Clotilde, La Réunion, France.,CNRS 9192, INSERM U1187, IRD 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Plateforme Technologique CYROI, Université de La Réunion, Sainte-Clotilde, La Réunion, France.,Laboratoire de Biologie, secteur : Laboratoire d'immunologie clinique et expérimentale ZOI (LICE OI), CHU La Réunion site Félix Guyon, St Denis, La Réunion, France
| |
Collapse
|
18
|
Bedoui Y, Neal JW, Gasque P. The Neuro-Immune-Regulators (NIREGs) Promote Tissue Resilience; a Vital Component of the Host's Defense Strategy against Neuroinflammation. J Neuroimmune Pharmacol 2018; 13:309-329. [PMID: 29909495 DOI: 10.1007/s11481-018-9793-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 01/29/2023]
Abstract
An effective protective inflammatory response in the brain is crucial for the clearance of pathogens (e.g. microbes, amyloid fibrils, prionSC) and should be closely regulated. However, the CNS seems to have limited tissue resilience to withstand the detrimental effects of uncontrolled inflammation compromising functional recovery and tissue repair. Newly described neuro-immune-regulators (NIREGs) are functionally related proteins regulating the severity and duration of the host inflammatory response. NIREGs such as CD200, CD47 and CX3CL1 are vital for increasing tissue resilience and are constitutively expressed by neurons. The interaction with co-receptors (CD200R, CD172a, CX3CR1) will maintain microglia in the resting phenotype, directing aggressive microglia phenotype and limiting bystander injuries. Neurons can also express many of the complement NIREGs (CD55, CD46, CD59 and factor H). Neurons and glia also express suppressor of cytokine signaling proteins (SOCS) down regulating janus kinase-signal transducer and activator of transcription (JAK/STAT) pathway and to lead to the polarization of microglia towards anti-inflammatory phenotype. Other NIREGs such as serine protease inhibitors (serpins) and thrombomodulin (CD141) inhibit neurotoxic systemic coagulation proteins such as thrombin. The unfolded protein response (UPR) detects misfolded proteins and other stressors to prevent irreversible cell injury. Microglial pattern recognition receptors (PRR) (TREM-2, CR3, FcγR) are important to clear apoptotic cells and cellular debris but in non-phlogystic manner through inhibitory signaling pathways. The TYRO3, Axl, Mer (TAM) tyrosine receptor kinases activated by Gas 6 and PROS1 regulate inflammation by inhibiting Toll like receptors (TLR) /JAK-STAT activation and contribute to NIREG's functions.
Collapse
Affiliation(s)
- Yosra Bedoui
- Université de la Réunion, CRNS 9192, INSERM U1187, IRD249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Plateforme Technologique CYROI, Saint -Clotilde, La Réunion, France
| | - Jim W Neal
- Infection and Immunity, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK.
| | - Philippe Gasque
- Laboratoire de biologie, secteur laboratoire d'immunologie Clinique et expérimentale ZOI, LICE-OI, CHU Felix Guyon Bellepierre, St Denis, La Réunion, France.
| |
Collapse
|
19
|
A role for viral infections in Parkinson's etiology? Neuronal Signal 2018; 2:NS20170166. [PMID: 32714585 PMCID: PMC7373231 DOI: 10.1042/ns20170166] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/06/2018] [Accepted: 03/19/2018] [Indexed: 02/06/2023] Open
Abstract
Despite over 200 years since its first description by James Parkinson, the cause(s) of most cases of Parkinson's disease (PD) are yet to be elucidated. The disparity between the current understanding of PD symptomology and pathology has led to numerous symptomatic therapies, but no strategy for prevention or disease cure. An association between certain viral infections and neurodegenerative diseases has been recognized, but largely ignored or dismissed as controversial, for decades. Recent epidemiological studies have renewed scientific interest in investigating microbial interactions with the central nervous system (CNS). This review examines past and current clinical findings and overviews the potential molecular implications of viruses in PD pathology.
Collapse
|
20
|
Calabrese V, Santoro A, Monti D, Crupi R, Di Paola R, Latteri S, Cuzzocrea S, Zappia M, Giordano J, Calabrese EJ, Franceschi C. Aging and Parkinson's Disease: Inflammaging, neuroinflammation and biological remodeling as key factors in pathogenesis. Free Radic Biol Med 2018; 115:80-91. [PMID: 29080843 DOI: 10.1016/j.freeradbiomed.2017.10.379] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/06/2017] [Accepted: 10/24/2017] [Indexed: 12/26/2022]
Abstract
In order to better understand the pathogenesis of Parkinson's Disease (PD) it is important to consider possible contributory factors inherent to the aging process, as age-related changes in a number of physiological systems (perhaps incurred within particular environments) appear to influence the onset and progression of neurodegenerative disorders. Accordingly, we posit that a principal mechanism underlying PD is inflammaging, i.e. the chronic inflammatory process characterized by an imbalance of pro- and anti-inflammatory mechanisms which has been recognized as operative in several age-related, and notably neurodegenerative diseases. Recent conceptualization suggests that inflammaging is part of the complex adaptive mechanisms ("re-modeling") that are ongoing through the lifespan, and which function to prevent or mitigate endogenous processes of tissue disruption and degenerative change(s). The absence of an adequate anti-inflammatory response can fuel inflammaging, which propagates on both local (i.e.- from cell to cell) and systemic levels (e.g.- via exosomes and other molecules present in the blood). In general, this scenario is compatible with the hypothesis that inflammaging represents a hormetic or hormetic-like effect, in which low levels of inflammatory stress may prompt induction of anti-inflammatory mediators and mechanisms, while sustained pro-inflammatory stress incurs higher and more durable levels of inflammatory substances, which, in turn prompt a local-to-systemic effect and more diverse inflammatory response(s). Given this perspective, new treatments of PD may be envisioned that strategically are aimed at exerting hormetic effects to sustain anti-inflammatory responses, inclusive perhaps, of modulating the inflammatory influence of the gut microbiota.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, via Santa Sofia 97, 95123 Catania, Italy; IBREGENS, Nutraceuticals and Functional Food Biotechnologies Research Associated, University of Catania, Italy.
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy; Interdepartmental Center "L. Galvani" (CIG), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Saverio Latteri
- Department of General Surgery, Cannizzaro Hospital, University of Catania, Catania, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Mario Zappia
- Department of Medical Sciences, Surgical and Advanced Technologies G.F. Ingrassia, Section of Neurosciences, University of Catania, Italy
| | - James Giordano
- Departments of Neurology and Biochemistry, and Neuroethics Studies Program, Georgetown University Medical Center, Washington, DC, USA
| | - Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, MA, USA
| | - Claudio Franceschi
- IRCCS, Institute of Neurological Sciences of Bologna, Via Altura 3, 40139 Bologna, Italy
| |
Collapse
|
21
|
Saeedi Saravi SS, Saeedi Saravi SS, Arefidoust A, Dehpour AR. The beneficial effects of HMG-CoA reductase inhibitors in the processes of neurodegeneration. Metab Brain Dis 2017; 32:949-965. [PMID: 28578514 DOI: 10.1007/s11011-017-0021-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Statins, cholesterol lowering drugs, have been demonstrated to exert beneficial effects in other conditions such as primary and progressing neurodegenerative diseases beyond their original role. Observation that statins ameliorate the neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS) and cerebral ischemic stroke, the neuroprotective effects of these drugs are thought to be linked to their anti-inflammatory, anti-oxidative, and anti-excitotoxic properties. Despite the voluminous literature on the clinical advantages of 3-hydroxy-3-methylglutaryl Co-enzyme A reductase (HMGCR) inhibitors (statins) in cardiovascular system, the neuroprotective effects and the underlying mechanisms are little understood. Hence, the present review tries to provide a critical overview on the statin-induced neuroprotection, which are presumed to be associated with the ability to reduce cholesterol, Amyloid-β and apolipoprotein E (ApoE) levels, decrease reactive oxygen and nitrogen species (ROS and RNS) formation, inhibit excitotoxicity, modulate matrix metalloproteinases (MMPs), stimulate endothelial nitric oxide synthase (eNOS), and increase cerebral blood perfusion. This review is also aimed to illustrate that statins protect neurons against the neuro-inflammatory processes through balancing pro-inflammatory/anti-inflammatory cytokines. Ultimately, the beneficial role of statins in ameliorating the development of PD, AD, MS and cerebral ischemic stroke has been separately reviewed.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Sobhan Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Arefidoust
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Wootla B, Denic A, Watzlawik JO, Warrington AE, Zoecklein LJ, Papke-Norton LM, David C, Rodriguez M. Human class I major histocompatibility complex alleles determine central nervous system injury versus repair. J Neuroinflammation 2016; 13:293. [PMID: 27855706 PMCID: PMC5112886 DOI: 10.1186/s12974-016-0759-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We investigated the role of human HLA class I molecules in persistent central nervous system (CNS) injury versus repair following virus infection of the CNS. METHODS Human class I A11+ and B27+ transgenic human beta-2 microglobulin positive (Hβ2m+) mice of the H-2 b background were generated on a combined class I-deficient (mouse beta-2 microglobulin deficient, β2m0) and class II-deficient (mouse Aβ0) phenotype. Intracranial infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL mice results in acute encephalitis with prominent injury in the hippocampus, striatum, and cortex. RESULTS Following infection with TMEV, a picornavirus, the Aβ0.β2m0 mice lacking active immune responses died within 18 to 21 days post-infection. These mice showed severe encephalomyelitis due to rapid replication of the viral genome. In contrast, transgenic Hβ2m mice with insertion of a single human class I MHC gene in the absence of human or mouse class II survived the acute infection. Both A11+ and B27+ mice significantly controlled virus RNA expression by 45 days and did not develop late-onset spinal cord demyelination. By 45 days post-infection (DPI), B27+ transgenic mice showed almost complete repair of the virus-induced brain injury, but A11+ mice conversely showed persistent severe hippocampal and cortical injury. CONCLUSIONS The findings support the hypothesis that the expression of a single human class I MHC molecule, independent of persistent virus infection, influences the extent of sub frequent chronic neuronal injury or repair in the absence of a class II MHC immune response.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Aleksandar Denic
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Jens O. Watzlawik
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224 USA
| | - Arthur E. Warrington
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Laurie J. Zoecklein
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Louisa M. Papke-Norton
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Chella David
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
23
|
Mao Y, Nguyen T, Sutherland T, Gorrie CA. Endogenous neural progenitor cells in the repair of the injured spinal cord. Neural Regen Res 2016; 11:1075-6. [PMID: 27630686 PMCID: PMC4994445 DOI: 10.4103/1673-5374.187035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Yilin Mao
- Neural Injury Research Unit, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Tara Nguyen
- Neural Injury Research Unit, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Theresa Sutherland
- Neural Injury Research Unit, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Catherine Anne Gorrie
- Neural Injury Research Unit, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
24
|
Zhou YQ, Liu Z, Liu HQ, Liu DQ, Chen SP, Ye DW, Tian YK. Targeting glia for bone cancer pain. Expert Opin Ther Targets 2016; 20:1365-1374. [PMID: 27428617 DOI: 10.1080/14728222.2016.1214716] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Bone cancer pain (BCP) remains to be a clinical challenge with limited pharmaceutical interventions. Therefore, novel therapeutic targets for the management of BCP are in desperate need. Recently, a growing body of evidence has suggested that glial cells may play a pivotal role in the pathogenesis of BCP. Areas covered: This review summarizes the recent progress in the understanding of glia in BCP and reveals the potential therapeutic targets in glia for BCP treatment. Expert opinion: Pharmacological interventions inhibiting the activation of glial cells, suppressing glia-derived proinflammatory cytokines, cell surface receptors, and the intracellular signaling pathways may be beneficial for the pain management of advanced cancer patients. However, these pharmacological interventions should not disrupt the normal function of glia cells since they play a vital supportive and protective role in the central nervous system.
Collapse
Affiliation(s)
- Ya-Qun Zhou
- a Research Center for Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China.,b Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Zheng Liu
- c Department of Urology , Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology , Wuhan , China
| | - Hui-Quan Liu
- d Cancer Center, Tongji Hospital, Tongji Medical college , Huazhong University of Science and Technology , Wuhan , China
| | - Dai-Qiang Liu
- a Research Center for Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China.,b Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Shu-Ping Chen
- a Research Center for Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China.,b Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| | - Da-Wei Ye
- d Cancer Center, Tongji Hospital, Tongji Medical college , Huazhong University of Science and Technology , Wuhan , China
| | - Yu-Ke Tian
- a Research Center for Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China.,b Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
25
|
Zhao B, Zou CJ, Zhou P. Delayed administration IL-1β neutralizing antibody improves cognitive function after transient global ischemia in rats. Behav Brain Res 2016; 303:53-60. [DOI: 10.1016/j.bbr.2016.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/06/2016] [Accepted: 01/10/2016] [Indexed: 12/23/2022]
|
26
|
Park HW, Chang JW, Yang YS, Oh W, Hwang JH, Kim DG, Paek SH. The Effect of Donor-Dependent Administration of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells following Focal Cerebral Ischemia in Rats. Exp Neurobiol 2015; 24:358-65. [PMID: 26713083 PMCID: PMC4688335 DOI: 10.5607/en.2015.24.4.358] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/27/2015] [Accepted: 10/29/2015] [Indexed: 01/02/2023] Open
Abstract
Stroke is an ischemic disease caused by clotted vessel-induced cell damage. It is characterized by high morbidity and mortality and is typically treated with a tissue plasminogen activator (tPA). However, this therapy is limited by temporal constraints. Recently, several studies have focused on cell therapy as an alternative treatment. Most researches have used fixed donor cell administration, and hence, the effect of donor-dependent cell administration is unknown. In this study, we administered 3 types of donor-derived human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) in the ischemic boundary zone of the ischemic stroke rat model. We then performed functional and pathological characterization using rotarod, the limb placement test, and immunofluorescent staining. We observed a significant decrease in neuron number, and notable stroke-like motor dysfunction, as assessed by the rotarod test (~40% decrease in time) and the limb placement test (4.5 point increase) in control rats with ischemic stroke. The neurobehavioral performance of the rats with ischemic stroke that were treated with hUCB-MSCs was significantly better than that of rats in the vehicle-injected control group. Regardless of which donor cells were used, hUCB-MSC transplantation resulted in an accumulation of neuronal progenitor cells, and angiogenic and tissue repair factors in the ischemic boundary zone. The neurogenic and angiogenic profiles of the 3 types of hUCB-MSCs were very similar. Our results suggest that intraparenchymal administration of hUCB-MSCs results in significant therapeutic effects in the ischemic brain regardless of the type of donor.
Collapse
Affiliation(s)
- Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Jong Wook Chang
- Stem Cell & Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul 13494, Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul 13494, Korea
| | - Jae Ha Hwang
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| |
Collapse
|
27
|
Gasque P, Jaffar-Bandjee MC. Chikungunya alphavirus infection in the nervous system and possible mechanisms of pathogenesis. Future Virol 2015. [DOI: 10.2217/fvl.15.88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chikungunya alphavirus (CHIKV) is transmitted by mosquitoes. Recent worldwide epidemics have revealed that CHIKV infects mainly muscles and joint tissues. Patients will suffer from arthralgia from months to years and leading to chronic arthritis. While CHIKV neuroinfection of susceptible hosts such as neonates is rare, it can lead to severe meningoencephalitis and encephalitis. CHIKV preferentially targets astrocytes, ependymal cells as well as epithelial cells of the choroid plexus. Remarkably, neurons are relatively spared but could suffer from the inflammatory response mounted by glial cells and recruited leukocytes. Interferon and related molecules as well as apoptosis will control the infectious challenge. Protective measures are critically needed and involving vaccination or passive immunization particularly in areas at risk of emerging epidemics.
Collapse
Affiliation(s)
- Philippe Gasque
- UMR PIMIT, Processus Infectieux en Milieu Insulaire Tropical, Université de la Réunion, INSERM 1187, CNRS 9192, IRD 249, Réunion Island
| | - Marie Christine Jaffar-Bandjee
- UMR PIMIT, Processus Infectieux en Milieu Insulaire Tropical, Université de la Réunion, INSERM 1187, CNRS 9192, IRD 249, Réunion Island
- Biology laboratory, CNR associé arbovirus, CHU Félix Guyon, Réunion Island
| |
Collapse
|
28
|
Borgmann K, Ghorpade A. HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads. Front Microbiol 2015; 6:1143. [PMID: 26579077 PMCID: PMC4621459 DOI: 10.3389/fmicb.2015.01143] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
As a popular psychostimulant, methamphetamine (METH) use leads to long-lasting, strong euphoric effects. While METH abuse is common in the general population, between 10 and 15% of human immunodeficiency virus-1 (HIV-1) patients report having abused METH. METH exacerbates the severity and onset of HIV-1-associated neurocognitive disorders (HAND) through direct and indirect mechanisms. Repetitive METH use impedes adherence to antiretroviral drug regimens, increasing the likelihood of HIV-1 disease progression toward AIDS. METH exposure also directly affects both innate and adaptive immunity, altering lymphocyte numbers and activity, cytokine signaling, phagocytic function and infiltration through the blood brain barrier. Further, METH triggers the dopamine reward pathway and leads to impaired neuronal activity and direct toxicity. Concurrently, METH and HIV-1 alter the neuroimmune balance and induce neuroinflammation, which modulates a wide range of brain functions including neuronal signaling and activity, glial activation, viral infection, oxidative stress, and excitotoxicity. Pathologically, reactive gliosis is a hallmark of both HIV-1- and METH-associated neuroinflammation. Significant commonality exists in the neurotoxic mechanisms for both METH and HAND; however, the pathways dysregulated in astroglia during METH exposure are less clear. Thus, this review highlights alterations in astrocyte intracellular signaling pathways, gene expression and function during METH and HIV-1 comorbidity, with special emphasis on HAND-associated neuroinflammation. Importantly, this review carefully evaluates interventions targeting astrocytes in HAND and METH as potential novel therapeutic approaches. This comprehensive overview indicates, without a doubt, that during HIV-1 infection and METH abuse, a complex dialog between all neural cells is orchestrated through astrocyte regulated neuroinflammation.
Collapse
Affiliation(s)
- Kathleen Borgmann
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| |
Collapse
|
29
|
Pastor JC, Rojas J, Pastor-Idoate S, Di Lauro S, Gonzalez-Buendia L, Delgado-Tirado S. Proliferative vitreoretinopathy: A new concept of disease pathogenesis and practical consequences. Prog Retin Eye Res 2015. [PMID: 26209346 DOI: 10.1016/j.preteyeres.2015.07.005] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During the last four decades, proliferative vitreoretinopathy (PVR) has defied the efforts of many researchers to prevent its occurrence or development. Thus, PVR is still the major complication following retinal detachment (RD) surgery and a bottle-neck for advances in cell therapy that require intraocular surgery. In this review we tried to combine basic and clinical knowledge, as an example of translational research, providing new and practical information for clinicians. PVR was defined as the proliferation of cells after RD. This idea was used for classifying PVR and also for designing experimental models used for testing many drugs, none of which were successful in humans. We summarize current information regarding the pathogenic events that follow any RD because this information may be the key for understanding and treating the earliest stages of PVR. A major focus is made on the intraretinal changes derived mainly from retinal glial cell reactivity. These responses can lead to intraretinal PVR, an entity that has not been clearly recognized. Inflammation is one of the major components of PVR, and we describe new genetic biomarkers that have the potential to predict its development. New treatment approaches are analyzed, especially those directed towards neuroprotection, which can also be useful for preventing visual loss after any RD. We also summarize the results of different surgical techniques and clinical information that is oriented toward the identification of high risk patients. Finally, we provide some recommendations for future classification of PVR and for designing comparable protocols for testing new drugs or techniques.
Collapse
Affiliation(s)
- J Carlos Pastor
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain.
| | - Jimena Rojas
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Universitario Austral, Universidad Austral, Buenos Aires, Argentina
| | - Salvador Pastor-Idoate
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Manchester Royal Eye Hospital, Manchester Vision Regeneration (MVR) Lab at NIHR/Wellcome Trust, Manchester, United Kingdom
| | - Salvatore Di Lauro
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
| | - Lucia Gonzalez-Buendia
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
| | - Santiago Delgado-Tirado
- Retina Group, IOBA (Eye Institute), University of Valladolid, Valladolid, Spain; Department of Ophthalmology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain
| |
Collapse
|
30
|
Hernangómez M, Carrillo-Salinas FJ, Mecha M, Correa F, Mestre L, Loría F, Feliú A, Docagne F, Guaza C. Brain innate immunity in the regulation of neuroinflammation: therapeutic strategies by modulating CD200-CD200R interaction involve the cannabinoid system. Curr Pharm Des 2015; 20:4707-22. [PMID: 24588829 PMCID: PMC4157566 DOI: 10.2174/1381612820666140130202911] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/29/2014] [Indexed: 11/24/2022]
Abstract
The central nervous system (CNS) innate immune response includes an arsenal of molecules and receptors expressed by professional phagocytes, glial cells and neurons that is involved in host defence and clearance of toxic and dangerous cell debris. However, any uncontrolled innate immune responses within the CNS are widely recognized as playing a major role in the development of autoimmune disorders and neurodegeneration, with multiple sclerosis (MS) Alzheimer's disease (AD) being primary examples. Hence, it is important to identify the key regulatory mechanisms involved in the control of CNS innate immunity and which could be harnessed to explore novel therapeutic avenues. Neuroimmune regulatory proteins (NIReg) such as CD95L, CD200, CD47, sialic acid, complement regulatory proteins (CD55, CD46, fH, C3a), HMGB1, may control the adverse immune responses in health and diseases. In the absence of these regulators, when neurons die by apoptosis, become infected or damaged, microglia and infiltrating immune cells are free to cause injury as well as an adverse inflammatory response in acute and chronic settings. We will herein provide new emphasis on the role of the pair CD200-CD200R in MS and its experimental models: experimental autoimmune encephalomyelitis (EAE) and Theiler’s virus induced demyelinating disease (TMEV-IDD). The interest of the cannabinoid system as inhibitor of inflammation prompt us to introduce our findings about the role of endocannabinoids (eCBs) in promoting CD200-CD200 receptor (CD200R) interaction and the benefits caused in TMEV-IDD. Finally, we also review the current data on CD200-CD200R interaction in AD, as well as, in the aging brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Carmen Guaza
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, 28002 Madrid, Spain.
| |
Collapse
|
31
|
Kashiwazaki H, Kakizaki M, Ikehara Y, Togayachi A, Narimatsu H, Watanabe R. Mice lacking α1,3-fucosyltransferase 9 exhibit modulation of in vivo immune responses against pathogens. Pathol Int 2015; 64:199-208. [PMID: 24888773 PMCID: PMC7167665 DOI: 10.1111/pin.12159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 03/25/2014] [Indexed: 01/13/2023]
Abstract
Carbohydrate structures, including Lewis X (Lex), which is not synthesized in mutant mice that lack α1,3‐fucosyltransferase 9 (Fut9−/−), are involved in cell–cell recognition and inflammation. However, immunological alteration in Fut9−/− mice has not been studied. Thus, the inflammatory response of Fut9−/− mice was examined using the highly neurovirulent mouse hepatitis virus (MHV) JHMV srr7 strain. Pathological study revealed that inflammation induced in the brains of Fut9−/− mice after infection was more extensive compared with that of wild‐type mice, although viral titers obtained from the brains of mutant mice were lower than those of wild‐type mice. Furthermore, the reduction in cell numbers in the spleens of wild‐type mice after infection was not observed in the infected Fut9−/− mice. Although there were no clear differences in the levels of cytokines examined in the brains between Fut9−/− and wild‐type mice except for interferon‐β (IFN‐β) expression, some of those in the spleens, including interferon‐γ (IFN‐γ), interleukin‐6 (IL‐6), and monocyte chemoattractant protein‐1 (MCP‐1), showed higher levels in Fut9−/− than in wild‐type mice. Furthermore, Fut9−/− mice were refractory to the in vivo inoculation of endotoxin (LPS) compared with wild‐type mice. These results indicate that Lex structures are involved in host responses against viral or bacterial challenges.
Collapse
Affiliation(s)
- Hiromi Kashiwazaki
- Department of BioinformaticsFaculty of EngineeringSoka UniversityHachiojiTokyoJapan
| | - Masatoshi Kakizaki
- Department of BioinformaticsFaculty of EngineeringSoka UniversityHachiojiTokyoJapan
| | - Yuzuru Ikehara
- Research Center for Medical GlycoscienceNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Akira Togayachi
- Research Center for Medical GlycoscienceNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Hisashi Narimatsu
- Research Center for Medical GlycoscienceNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Rihito Watanabe
- Department of BioinformaticsFaculty of EngineeringSoka UniversityHachiojiTokyoJapan
| |
Collapse
|
32
|
Das T, Hoarau JJ, Bandjee MCJ, Maquart M, Gasque P. Multifaceted innate immune responses engaged by astrocytes, microglia and resident dendritic cells against Chikungunya neuroinfection. J Gen Virol 2014; 96:294-310. [PMID: 25351727 DOI: 10.1099/vir.0.071175-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chikungunya virus (CHIKV) has recently affected millions of people in the Indian Ocean, with rare cases of encephalopathy and encephalitis occurring in neonates. In the study described herein, the capacity of mouse brain cells to control infection through innate immune antiviral responses was assessed. In vitro, CHIKV principally infected a subpopulation of mouse GFAP+ primary astrocytes. Oligodendrocytes and neurons could also be infected. An innate immune response was engaged by CHIKV-infected astrocytes with elevated expression of mRNAs for IFN-α-β, inflammatory cytokines (e.g. IL-1β, IL-12, IL-10, IL-24) and proapoptotic factors (e.g. TNF-α, FasL, Lymphotoxin B). Programmed cell death through the intrinsic caspase-9 pathway was observed by immunofluorescence in infected astrocytes and neurons but not in oligodendrocytes. Interestingly, microglia did not replicate CHIKV but responded by elevated mitogen-activated protein kinase (MAPK) activity. Intracerebroventricular injection of CHIKV in neonate mice led to the infection of astrocytes. The astrogliosis response was accompanied by a dendritic CD206+ cell mobilization restricted to the site of infection. The results of this study support the paradigm that a multifaceted innate immune response can be mobilized by both professional immune and glial cells to control CHIKV neuroinfection events in neonates.
Collapse
Affiliation(s)
- Trina Das
- Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| | - Jean Jacques Hoarau
- Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| | - Marie Christine Jaffar Bandjee
- Virology laboratory, CHU Félix Guyon of la Réunion, St Denis, Reunion Island.,Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| | - Marianne Maquart
- CIRAD and CYROI, St Denis, Reunion Island.,CRVOI, St Denis, Reunion Island
| | - Philippe Gasque
- Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| |
Collapse
|
33
|
Method of labelling of individual ependymal areas according to periventricular structures of the rat lateral brain ventricles. Biologia (Bratisl) 2014. [DOI: 10.2478/s11756-014-0421-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
34
|
Gérardin P, Sampériz S, Ramful D, Boumahni B, Bintner M, Alessandri JL, Carbonnier M, Tiran-Rajaoefera I, Beullier G, Boya I, Noormahomed T, Okoï J, Rollot O, Cotte L, Jaffar-Bandjee MC, Michault A, Favier F, Kaminski M, Fourmaintraux A, Fritel X. Neurocognitive outcome of children exposed to perinatal mother-to-child Chikungunya virus infection: the CHIMERE cohort study on Reunion Island. PLoS Negl Trop Dis 2014; 8:e2996. [PMID: 25033077 PMCID: PMC4102444 DOI: 10.1371/journal.pntd.0002996] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 05/22/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Little is known about the neurocognitive outcome in children exposed to perinatal mother-to-child Chikungunya virus (p-CHIKV) infection. METHODS The CHIMERE ambispective cohort study compared the neurocognitive function of 33 p-CHIKV-infected children (all but one enrolled retrospectively) at around two years of age with 135 uninfected peers (all enrolled prospectively). Psychomotor development was assessed using the revised Brunet-Lezine scale, examiners blinded to infectious status. Development quotients (DQ) with subscores covering movement/posture, coordination, language, sociability skills were calculated. Predictors of global neurodevelopmental delay (GND, DQ ≤ 85), were investigated using multivariate Poisson regression modeling. Neuroradiologic follow-up using magnetic resonance imaging (MRI) scans was proposed for most of the children with severe forms. RESULTS The mean DQ score was 86.3 (95%CI: 81.0-91.5) in infected children compared to 100.2 (95%CI: 98.0-102.5) in uninfected peers (P<0.001). Fifty-one percent (n = 17) of infected children had a GND compared to 15% (n = 21) of uninfected children (P<0.001). Specific neurocognitive delays in p-CHIKV-infected children were as follows: coordination and language (57%), sociability (36%), movement/posture (27%). After adjustment for maternal social situation, small for gestational age, and head circumference, p-CHIKV infection was found associated with GND (incidence rate ratio: 2.79, 95%CI: 1.45-5.34). Further adjustments on gestational age or breastfeeding did not change the independent effect of CHIKV infection on neurocognitive outcome. The mean DQ of p-CHIKV-infected children was lower in severe encephalopathic children than in non-severe children (77.6 versus 91.2, P<0.001). Of the 12 cases of CHIKV neonatal encephalopathy, five developed a microcephaly (head circumference <-2 standard deviations) and four matched the definition of cerebral palsy. MRI scans showed severe restrictions of white matter areas, predominant in the frontal lobes in these children. CONCLUSIONS The neurocognitive outcome of children exposed to perinatal mother-to-child CHIKV infection is poor. Severe CHIKV neonatal encephalopathy is associated with an even poorer outcome.
Collapse
Affiliation(s)
- Patrick Gérardin
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
- INSERM CIC-EC (CIE2), Saint-Pierre, La Réunion, France
- INSERM UMRS 953, “Epidemiological Research Unit on Perinatal Health and Women and Children Health”, UPMC Université Paris 6, Paris, France
| | | | - Duksha Ramful
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
- INSERM CIC-EC (CIE2), Saint-Pierre, La Réunion, France
- GRI, Research Group on Immunopathology and Infection, EA4517, Université de La Réunion, INSERM UMRS 945 “Immunity and Infection” Saint-Denis, La Réunion, France
| | - Brahim Boumahni
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | - Marc Bintner
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | | | | | | | - Gilles Beullier
- Centre Hospitalier Gabriel Martin, Saint-Paul, La Réunion, France
| | - Irénée Boya
- Centre Hospitalier de l'Est Réunion, Saint-Benoît, La Réunion, France
| | | | - Jocelyn Okoï
- Clinique Durieux, Le Tampon, La Réunion, France
- Centre d'Action Médico-Sociale Précoce (CAMSP), Saint-Louis, La Réunion, France
| | | | - Liliane Cotte
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | | | - Alain Michault
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | | | - Monique Kaminski
- INSERM UMRS 953, “Epidemiological Research Unit on Perinatal Health and Women and Children Health”, UPMC Université Paris 6, Paris, France
| | | | - Xavier Fritel
- INSERM UMRS 953, “Epidemiological Research Unit on Perinatal Health and Women and Children Health”, UPMC Université Paris 6, Paris, France
- Poitiers University Hospital, Poitiers, France
- INSERM CIC-P 0802, Poitiers, France
| |
Collapse
|
35
|
Asea A, Kaur P, Panossian A, Wikman KG. Evaluation of molecular chaperons Hsp72 and neuropeptide Y as characteristic markers of adaptogenic activity of plant extracts. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:1323-1329. [PMID: 23920279 DOI: 10.1016/j.phymed.2013.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 05/28/2013] [Accepted: 07/02/2013] [Indexed: 06/02/2023]
Abstract
We have previously demonstrated that ADAPT-232, a fixed combination of adaptogenic substances derived from Eleutherococcus senticosus root extract, Schisandra chinensis berry extract, Rhodiola rosea root extract stimulated the expression and release of neuropeptide Y (NPY) and molecular chaperone Hsp72 from isolated human neurolgia cells. Both of these mediators of stress response are known to play an important role in regulation of neuroendocrine system and immune response. We further demonstrated that ADAPT-232 induced release of Hsp70 is mediated by NPY, suggesting an existence of NPY-mediated pathway of activation of Hsp72 release into the blood circulation system. The objective of this study was to determine whether this pathway is common for adaptogens and whether NPY and/or Hsp72 can be considered as necessary specific biomarkers for adaptogenic activity. The release of NPY and Hsp72 from neuroglia cells in response to treatment with various plant extracts (n=23) including selected validated adaptogens, partly validated adaptogens, claimed but negligibly validated adaptogens and some other plant extracts affecting neuroendocrine and immune systems but never considered as adaptogens was measured using high throughput ELISA techniques. We demonstrated that adaptogens, e.g. R. rosea, S. chinensis and E. senticosus stimulate both NPY and Hsp70 release from neuroblastoma cells, while tonics and stimulants have no significant effect on NPY in this in vitro test. In the groups of partly validated adaptogens the effect of Panax ginseng and Withania somnifera was not statistically significant both on NPY and Hsp70 release, while the activating effect of Bryonia alba and Rhaponticum cartamoides was significant only on Hsp70. In contrast, all tested non-adaptogens, such as antiinflammatoty plant extracts Matricaria recutita, Pelargonium sidoides, Hedera helix and Vitis vinifera significantly inhibit Hsp70 release and have no influence on NPY release from neuroblastoma cells. These experiments were further validated using primary human neurons and confirmed that adaptogens activate the release of both NPY and Hsp70, while tested non adaptogens were inactive in NPY assay and inhibit the release of Hsp70. Taken together, our data demonstrates for the first time that neuropeptide Y and heat shock protein Hsp70 can be used as molecular biomarkers for adaptogenic activity.
Collapse
Affiliation(s)
- Alexzander Asea
- Morehouse School of Medicine, Department of Microbiology, Biochemistry and Immunology & Department of Pathology and Laboratory Medicine, 720 Westview Drive SW, Atlanta, GA 30310-1495, USA.
| | | | | | | |
Collapse
|
36
|
Ricci S, Gerlini A, Pammolli A, Chiavolini D, Braione V, Tripodi SA, Colombari B, Blasi E, Oggioni MR, Peppoloni S, Pozzi G. Contribution of different pneumococcal virulence factors to experimental meningitis in mice. BMC Infect Dis 2013; 13:444. [PMID: 24059458 PMCID: PMC3848944 DOI: 10.1186/1471-2334-13-444] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/13/2013] [Indexed: 11/28/2022] Open
Abstract
Background Pneumococcal meningitis (PM) is a life-threatening disease with a high case-fatality rate and elevated risk for serious neurological sequelae. In this study, we investigated the contribution of three major virulence factors of Streptococcus pneumoniae, the capsule, pneumococcal surface protein A (PspA) and C (PspC), to the pathogenesis of experimental PM. Methods Mice were challenged by the intracranial route with the serotype 4 TIGR4 strain (wt) and three isogenic mutants devoid of PspA, PspC, and the capsule. Survival, bacterial counts, and brain histology were carried out. To study the interaction between S. pneumoniae mutants and microglia, phagocytosis and survival experiments were performed using the BV2 mouse microglial cell line. Results Virulence of the PspC mutant was comparable to that of TIGR4. In contrast, survival of animals challenged with the PspA mutant was significantly increased compared with the wt, and the mutant was also impaired at replicating in the brain and blood of infected mice. Brain histology indicated that all strains, except for the unencapsulated mutant, caused PM. Analysis of inflammation and damage in the brain of mice infected with TIGR4 or its unencapsulated mutant demonstrated that the rough strain was unable to induce inflammation and neuronal injury, even at high challenge doses. Results with BV2 cells showed no differences in phagocytic uptake between wt and mutants. In survival assays, however, the PspA mutant showed significantly reduced survival in microglia compared with the wt. Conclusions PspA contributed to PM pathogenesis possibly by interacting with microglia at early infection stages, while PspC had limited importance in the disease. The rough mutant did not cause brain inflammation, neuronal damage or mouse death, strengthening the key role of the capsule in PM.
Collapse
Affiliation(s)
- Susanna Ricci
- Department of Medical Biotechnologies, Laboratory of Molecular Microbiology and Biotechnology (LA,M,M,B,), University of Siena and Siena University Hospital, Siena 53100, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ramesh G, MacLean AG, Philipp MT. Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain. Mediators Inflamm 2013; 2013:480739. [PMID: 23997430 PMCID: PMC3753746 DOI: 10.1155/2013/480739] [Citation(s) in RCA: 422] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/11/2013] [Accepted: 07/12/2013] [Indexed: 01/18/2023] Open
Abstract
Cytokines and chemokines are proteins that coordinate the immune response throughout the body. The dysregulation of cytokines and chemokines is a central feature in the development of neuroinflammation, neurodegeneration, and demyelination both in the central and peripheral nervous systems and in conditions of neuropathic pain. Pathological states within the nervous system can lead to activation of microglia. The latter may mediate neuronal and glial cell injury and death through production of proinflammatory factors such as cytokines and chemokines. These then help to mobilize the adaptive immune response. Although inflammation may induce beneficial effects such as pathogen clearance and phagocytosis of apoptotic cells, uncontrolled inflammation can result in detrimental outcomes via the production of neurotoxic factors that exacerbate neurodegenerative pathology. In states of prolonged inflammation, continual activation and recruitment of effector cells can establish a feedback loop that perpetuates inflammation and ultimately results in neuronal injury. A critical balance between repair and proinflammatory factors determines the outcome of a neurodegenerative process. This review will focus on how cytokines and chemokines affect neuroinflammation and disease pathogenesis in bacterial meningitis and brain abscesses, Lyme neuroborreliosis, human immunodeficiency virus encephalitis, and neuropathic pain.
Collapse
Affiliation(s)
- Geeta Ramesh
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703 Three Rivers Road, Covington, LA 70433, USA.
| | | | | |
Collapse
|
38
|
How does the brain limit the severity of inflammation and tissue injury during bacterial meningitis? J Neuropathol Exp Neurol 2013; 72:370-85. [PMID: 23584204 DOI: 10.1097/nen.0b013e3182909f2f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The most devastating CNS bacterial infection, bacterial meningitis, has both acute and long-term neurologic consequences. The CNS defends itself against bacterial invasion through a combination of physical barriers (i.e. blood-brain barrier, meninges, and ependyma), which contain macrophages that express a range of pattern-recognition receptors that detect pathogens before they gain access to the CNS and cerebrospinal fluid. This activates an antipathogen response consisting of inflammatory cytokines, complement, and chemoattractants. Regulation of the antipathogen inflammatory response is essential for preventing irreversible brain injury and protecting stem cell populations in the ventricle wall. The severity of brain inflammation is regulated by the clearance of apoptotic inflammatory cells and neurons. Death signaling pathways are expressed by glia to stimulate apoptosis of neutrophils, lymphocytes, and damaged neurons and to regulate in flammation and remove necrotic cells. The emerging group of neuroimmunoregulatory molecules adjusts the balance of the anti-inflammatory and proinflammatory response to provide optimal conditions for effective clearance of pathogens and apoptotic cells but reduce the severity of the inflammatory response to prevent injury to brain cells, including stem cell populations. The neuroimmunoregulatory molecules and other CNS anti-inflammatory pathways represent potential therapeutic targets capable of reducing brain injury caused by bacterial infection.
Collapse
|
39
|
Renner NA, Sansing HA, Inglis FM, Mehra S, Kaushal D, Lackner AA, Maclean AG. Transient acidification and subsequent proinflammatory cytokine stimulation of astrocytes induce distinct activation phenotypes. J Cell Physiol 2013; 228:1284-94. [PMID: 23154943 DOI: 10.1002/jcp.24283] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 11/02/2012] [Indexed: 01/18/2023]
Abstract
The foot processes of astrocytes cover over 60% of the surface of brain microvascular endothelial cells, regulating tight junction integrity. Retraction of astrocyte foot processes has been postulated to be a key mechanism in pathology. Therefore, movement of an astrocyte in response to a proinflammatory cytokine or even limited retraction of processes would result in leaky junctions between endothelial cells. Astrocytes lie at the gateway to the CNS and are instrumental in controlling leukocyte entry. Cultured astrocytes typically have a polygonal morphology until stimulated. We hypothesized that cultured astrocytes which were induced to stellate would have an activated phenotype compared with polygonal cells. We investigated the activation of astrocytes derived from adult macaques to the cytokine TNF-α under resting and stellated conditions by four parameters: morphology, intermediate filament expression, adhesion, and cytokine secretion. Astrocytes were stellated following transient acidification; resulting in increased expression of GFAP and vimentin. Stellation was accompanied by decreased adhesion that could be recovered with proinflammatory cytokine treatment. Surprisingly, there was decreased secretion of proinflammatory cytokines by stellated astrocytes compared with polygonal cells. These results suggest that astrocytes are capable of multiple phenotypes depending on the stimulus and the order stimuli are applied.
Collapse
Affiliation(s)
- Nicole A Renner
- Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Peppoloni S, Colombari B, Beninati C, Felici F, Teti G, Speziale P, Ricci S, Ardizzoni A, Manca L, Blasi E. The Spr1875 protein confers resistance to the microglia-mediated killing of Streptococcus pneumoniae. Microb Pathog 2013; 59-60:42-7. [PMID: 23587464 DOI: 10.1016/j.micpath.2013.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/27/2013] [Accepted: 04/03/2013] [Indexed: 12/11/2022]
Abstract
By screening a whole-genome λ-display library of Streptococcus pneumoniae, we have previously identified a novel surface protein, named Spr1875, that exhibited immunogenic properties and was closely related to pneumococcal virulence. In the present study, we investigated the role of the Spr1875 antigen in the interaction of S. pneumoniae with microglia, the resident brain macrophages. By using an in vitro infection model, the BV2 microglial cell line was challenged with the S. pneumoniae strain DP1004 and its isogenic spr1875-deleted mutant (Δspr1875). Both strains were phagocytosed by microglia efficiently and to a similar extent; however, the DP1004 strain was more resistant than the Δspr1875 mutant to the intracellular killing, as assessed by antibiotic protection and phagosome maturation assays. Moreover, significant differences between the two strains were also observed in terms of susceptibility to microglia-mediated killing. Taken together, these results indicate that S. pneumoniae-microglial cell interplay is influenced by the presence of Spr1875, suggesting that this protein may play a role in the pathogenesis of pneumococcal meningitis.
Collapse
Affiliation(s)
- Samuele Peppoloni
- Dipartimento di Medicina Diagnostica, Clinica e di Sanità Pubblica, Università di Modena e Reggio Emilia (Unimore), 41125 Modena, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder characterized by the aggregation of two proteins, amyloid-β and hyperphosphorylated tau, and by neuronal and synaptic loss. Although some drugs have been shown to slow the progression of the disease, at present no treatment has been developed that can stop or reverse the progression of the pathology. Recently, new therapeutic strategies have been proposed for the treatment of the disease. Among these, the development of stem cells and gene-modified cells is an especially promising therapeutic approach for AD. In this review we highlight the experimental and preclinical studies that have been focused on stem cell-based and gene-modified cell-based uses as potential therapies for AD. The potential clinical applications are also discussed.
Collapse
Affiliation(s)
- Micaela Johanna Glat
- Laboratory of Neuroscience, Sackler Faculty of Medicine, Felsenstein Medical Research Center, Tel Aviv University, Petah-Tikva, Israel
| | | |
Collapse
|
42
|
Panossian A, Hamm R, Kadioglu O, Wikman G, Efferth T. Synergy and Antagonism of Active Constituents of ADAPT-232 on Transcriptional Level of Metabolic Regulation of Isolated Neuroglial Cells. Front Neurosci 2013; 7:16. [PMID: 23430930 PMCID: PMC3576868 DOI: 10.3389/fnins.2013.00016] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/01/2013] [Indexed: 12/16/2022] Open
Abstract
Gene expression profiling was performed on the human neuroglial cell line T98G after treatment with adaptogen ADAPT-232 and its constituents – extracts of Eleutherococcus senticosus root, Schisandra chinensis berry, and Rhodiola rosea root as well as several constituents individually, namely, eleutheroside E, schizandrin B, salidroside, triandrin, and tyrosol. A common feature for all tested adaptogens was their effect on G-protein-coupled receptor signaling pathways, i.e., cAMP, phospholipase C (PLC), and phosphatidylinositol signal transduction pathways. Adaptogens may reduce the cAMP level in brain cells by down-regulation of adenylate cyclase gene ADC2Y and up-regulation of phosphodiesterase gene PDE4D that is essential for energy homeostasis as well as for switching from catabolic to anabolic states and vice versa. Down-regulation of cAMP by adaptogens may decrease cAMP-dependent protein kinase A activity in various cells resulting in inhibition stress-induced catabolic transformations and saving of ATP for many ATP-dependant metabolic transformations. All tested adaptogens up-regulated the PLCB1 gene, which encodes phosphoinositide-specific PLC and phosphatidylinositol 3-kinases (PI3Ks), key players for the regulation of NF-κB-mediated defense responses. Other common targets of adaptogens included genes encoding ERα estrogen receptor (2.9–22.6 fold down-regulation), cholesterol ester transfer protein (5.1–10.6 fold down-regulation), heat shock protein Hsp70 (3.0–45.0 fold up-regulation), serpin peptidase inhibitor (neuroserpin), and 5-HT3 receptor of serotonin (2.2–6.6 fold down-regulation). These findings can be reconciled with the observed beneficial effects of adaptogens in behavioral, mental, and aging-associated disorders. Combining two or more active substances in one mixture significantly changes deregulated genes profiles: synergetic interactions result in activation of genes that none of the individual substances affected, while antagonistic interactions result in suppression some genes activated by individual substances. These interactions can have an influence on transcriptional control of metabolic regulation both on the cellular level and the level of the whole organism. Merging of deregulated genes array profiles and intracellular networks is specific to the new substance with unique pharmacological characteristics. Presumably, this phenomenon could be used to eliminate undesirable effects (e.g., toxic effects) and increase the selectivity of pharmacological intervention.
Collapse
|
43
|
Abstract
Metabolic syndrome, a network of medical disorders that greatly increase the risk for developing metabolic and cardiovascular diseases, has reached epidemic levels in many areas of today's world. Despite this alarming medicare situation, scientific understandings on the root mechanisms of metabolic syndrome are still limited, and such insufficient knowledge contributes to the relative lack of effective treatments or preventions for related diseases. Recent interdisciplinary studies from neuroendocrinology and neuroimmunology fields have revealed that overnutrition can trigger intracellular stresses to cause inflammatory changes mediated by molecules that control innate immunity. This type of nutrition-related molecular inflammation in the central nervous system, particularly in the hypothalamus, can form a common pathogenic basis for the induction of various metabolic syndrome components such as obesity, insulin resistance, and hypertension. Proinflammatory NF-κB pathway has been revealed as a key molecular system for pathologic induction of brain inflammation, which translates overnutrition and resulting intracellular stresses into central neuroendocrine and neural dysregulations of energy, glucose, and cardiovascular homeostasis, collectively leading to metabolic syndrome. This article reviews recent research advances in the neural mechanisms of metabolic syndrome and related diseases from the perspective of pathogenic induction by intracellular stresses and NF-κB pathway of the brain.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Molecular Pharmacology and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
44
|
Combes V, Guillemin GJ, Chan-Ling T, Hunt NH, Grau GER. The crossroads of neuroinflammation in infectious diseases: endothelial cells and astrocytes. Trends Parasitol 2012; 28:311-9. [PMID: 22727810 DOI: 10.1016/j.pt.2012.05.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 05/29/2012] [Accepted: 05/29/2012] [Indexed: 01/06/2023]
Abstract
Homeostasis implies constant operational defence mechanisms, against both external and internal threats. Infectious agents are prominent among such threats. During infection, the host elicits the release of a vast array of molecules and numerous cell-cell interactions are triggered. These pleiomorphic mediators and cellular effects are of prime importance in the defence of the host, both in the systemic circulation and at sites of tissue injury, for example, the blood-brain barrier (BBB). Here, we focus on the interactions between the endothelium, astrocytes, and the molecules they release. Our review addresses these interactions during infectious neurological diseases of various origins, especially cerebral malaria (CM). Two novel elements of the interplay between endothelium and astrocytes, microparticles and the kynurenine pathway, will also be discussed.
Collapse
Affiliation(s)
- Valéry Combes
- Vascular Immunology Unit, Sydney Medical School and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | | | | | | | | |
Collapse
|
45
|
Abstract
The hypothalamus is one of the master regulators of various physiological processes, including energy balance and nutrient metabolism. These regulatory functions are mediated by discrete hypothalamic regions that integrate metabolic sensing with neuroendocrine and neural controls of systemic physiology. Neurons and nonneuronal cells in these hypothalamic regions act supportively to execute metabolic regulations. Under conditions of brain and hypothalamic inflammation, which may result from overnutrition-induced intracellular stresses or disease-associated systemic inflammatory factors, extracellular and intracellular environments of hypothalamic cells are disrupted, leading to central metabolic dysregulations and various diseases. Recent research has begun to elucidate the effects of hypothalamic inflammation in causing diverse components of metabolic syndrome leading to diabetes and cardiovascular disease. These new understandings have provocatively expanded previous knowledge on the cachectic roles of brain inflammatory response in diseases, such as infections and cancers. This review describes the molecular and cellular characteristics of hypothalamic inflammation in metabolic syndrome and related diseases as opposed to cachectic diseases, and also discusses concepts and potential applications of inhibiting central/hypothalamic inflammation to treat nutritional diseases.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Molecular Pharmacology, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | |
Collapse
|
46
|
Denizot M, Neal JW, Gasque P. Encephalitis due to emerging viruses: CNS innate immunity and potential therapeutic targets. J Infect 2012; 65:1-16. [PMID: 22484271 DOI: 10.1016/j.jinf.2012.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 02/27/2012] [Accepted: 03/08/2012] [Indexed: 12/21/2022]
Abstract
The emerging viruses represent a group of pathogens that are intimately connected to a diverse range of animal vectors. The recent escalation of air travel climate change and urbanization has meant humans will have increased risk of contacting these pathogens resulting in serious CNS infections. Many RNA viruses enter the CNS by evading the BBB due to axonal transport from the periphery. The systemic adaptive and CNS innate immune systems express pattern recognition receptors PRR (TLRs, RiG-1 and MDA-5) that detect viral nucleic acids and initiate host antiviral response. However, several emerging viruses (West Nile Fever, Influenza A, Enterovirus 71 Ebola) are recognized and internalized by host cell receptors (TLR, MMR, DC-SIGN, CD162 and Scavenger receptor B) and escape immuno surveillance by the host systemic and innate immune systems. Many RNA viruses express viral proteins WNF (E protein), Influenza A (NS1), EV71 (protein 3C), Rabies (Glycoprotein), Ebola proteins (VP24 and VP 35) that inhibit the host cell anti-virus Interferon type I response promoting virus replication and encephalitis. The therapeutic use of RNA interference methodologies to silence gene expression of viral peptides and treat emerging virus infection of the CNS is discussed.
Collapse
Affiliation(s)
- M Denizot
- GRI, Immunopathology and Infectious Disease Research Grouping (IRG, GRI), University of La Reunion, Reunion
| | | | | |
Collapse
|
47
|
Ransohoff RM, Brown MA. Innate immunity in the central nervous system. J Clin Invest 2012; 122:1164-71. [PMID: 22466658 DOI: 10.1172/jci58644] [Citation(s) in RCA: 716] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Immune responses in the CNS are common, despite its perception as a site of immune privilege. These responses can be mediated by resident microglia and astrocytes, which are innate immune cells without direct counterparts in the periphery. Furthermore, CNS immune reactions often take place in virtual isolation from the innate/adaptive immune interplay that characterizes peripheral immunity. However, microglia and astrocytes also engage in significant cross-talk with CNS-infiltrating T cells and other components of the innate immune system. Here we review the cellular and molecular basis of innate immunity in the CNS and discuss what is known about how outcomes of these interactions can lead to resolution of infection, neurodegeneration, or neural repair depending on the context.
Collapse
Affiliation(s)
- Richard M Ransohoff
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
48
|
Van Goethem E, Silva EA, Xiao H, Franc NC. The Drosophila TRPP cation channel, PKD2 and Dmel/Ced-12 act in genetically distinct pathways during apoptotic cell clearance. PLoS One 2012; 7:e31488. [PMID: 22347485 PMCID: PMC3275576 DOI: 10.1371/journal.pone.0031488] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 01/09/2012] [Indexed: 11/18/2022] Open
Abstract
Apoptosis, a genetically programmed cell death, allows for homeostasis and tissue remodelling during development of all multi-cellular organisms. Phagocytes swiftly recognize, engulf and digest apoptotic cells. Yet, to date the molecular mechanisms underlying this phagocytic process are still poorly understood. To delineate the molecular mechanisms of apoptotic cell clearance in Drosophila, we have carried out a deficiency screen and have identified three overlapping phagocytosis-defective mutants, which all delete the fly homologue of the ced-12 gene, known as Dmel\ced12. As anticipated, we have found that Dmel\ced-12 is required for apoptotic cell clearance, as for its C. elegans and mammalian homologues, ced-12 and elmo, respectively. However, the loss of Dmel\ced-12 did not solely account for the phenotypes of all three deficiencies, as zygotic mutations and germ line clones of Dmel\ced-12 exhibited weaker phenotypes. Using a nearby genetically interacting deficiency, we have found that the polycystic kidney disease 2 gene, pkd2, which encodes a member of the TRPP channel family, is also required for phagocytosis of apoptotic cells, thereby demonstrating a novel role for PKD2 in this process. We have also observed genetic interactions between pkd2, simu, drpr, rya-r44F, and retinophilin (rtp), also known as undertaker (uta), a gene encoding a MORN-repeat containing molecule, which we have recently found to be implicated in calcium homeostasis during phagocytosis. However, we have not found any genetic interaction between Dmel\ced-12 and simu. Based on these genetic interactions and recent reports demonstrating a role for the mammalian pkd-2 gene product in ER calcium release during store-operated calcium entry, we propose that PKD2 functions in the DRPR/RTP pathway to regulate calcium homeostasis during this process. Similarly to its C. elegans homologue, Dmel\Ced-12 appears to function in a genetically distinct pathway.
Collapse
Affiliation(s)
- Emeline Van Goethem
- Medical Research Council Cell Biology Unit, MRC Laboratory for Molecular Cell Biology and Anatomy and Developmental Biology Department, University College London, London, United Kingdom
| | - Elizabeth A. Silva
- Medical Research Council Cell Biology Unit, MRC Laboratory for Molecular Cell Biology and Anatomy and Developmental Biology Department, University College London, London, United Kingdom
| | - Hui Xiao
- The Department of Genetics, Affiliated to the Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, California, United States of America
| | - Nathalie C. Franc
- The Department of Genetics, Affiliated to the Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
Pfrieger FW, Slezak M. Genetic approaches to study glial cells in the rodent brain. Glia 2011; 60:681-701. [PMID: 22162024 DOI: 10.1002/glia.22283] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 11/18/2011] [Indexed: 01/02/2023]
Abstract
The development, function, and pathology of the brain depend on interactions of neurons and different types of glial cells, namely astrocytes, oligodendrocytes, microglia, and ependymal cells. Understanding neuron-glia interactions in vivo requires dedicated experimental approaches to manipulate each cell type independently. In this review, we first summarize techniques that allow for cell-specific gene modification including targeted mutagenesis and viral transduction. In the second part, we describe the genetic models that allow to target the main glial cell types in the central nervous system. The existing arsenal of approaches to study glial cells in vivo and its expansion in the future are key to understand neuron-glia interactions under normal and pathologic conditions.
Collapse
Affiliation(s)
- Frank W Pfrieger
- CNRS UPR 3212, University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI), 67084 Strasbourg, France.
| | | |
Collapse
|
50
|
Graham SP, Earley RL, Guyer C, Mendonça MT. Innate immune performance and steroid hormone profiles of pregnant versus nonpregnant cottonmouth snakes (Agkistrodon piscivorus). Gen Comp Endocrinol 2011; 174:348-53. [PMID: 21986088 DOI: 10.1016/j.ygcen.2011.09.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 09/22/2011] [Accepted: 09/25/2011] [Indexed: 11/25/2022]
Abstract
Squamates (lizards and snakes) have independently evolved viviparity over 100 times, and exhibit a wide range of maternal investment in developing embryos from the extremes of lecithotrophic oviparity to matrotrophic viviparity. This group therefore provides excellent comparative opportunities for studying endocrine and immune involvement during pregnancy, and their possible interactions. We studied the cottonmouth (Agkistrodon piscivorus), since they exhibit limited placentation (e.g., ovoviviparity), allowing comparison with squamate species hypothesized to require considerable maternal immune modulation due to the presence of a more extensive placental connection. Furthermore, the cottonmouth's biennial reproductive cycle provides an opportunity for simultaneously comparing pregnant and non-pregnant females in the wild. We document significantly elevated concentrations of progesterone (P4) and significantly lower concentrations of estradiol (E2) in pregnant females relative to non-pregnant females. Pregnant females had lower plasma bacteria lysis capacity relative to non-pregnant females. This functional measure of innate immunity is a proxy for complement performance, and we also determined significant correlations between P4 and decreased complement performance in pregnant females. These findings are consistent with studies that have determined P4's role in complement modulation during pregnancy in mammals, and thus this study joins a growing number of studies that have demonstrated convergent and/or conserved physiological mechanisms regulating viviparous reproduction in vertebrates.
Collapse
Affiliation(s)
- Sean P Graham
- Department of Biological Sciences, 331 Funchess Hall, Auburn University, Auburn, AL 36849, USA.
| | | | | | | |
Collapse
|