1
|
Graindorge PH, Paoli J, Yildirim B, Morel C, Herzine A, Collin M, Gallais I, Boucard S, Pouyatos B, Meyre D, Lagadic-Gossmann D, Sergent O, Schroeder H, Grova N. Early stage of metabolic dysfunction associated steatotic liver disease disrupts circadian rhythm and induces neuroinflammation in rats. Sci Rep 2025; 15:10616. [PMID: 40148513 PMCID: PMC11950343 DOI: 10.1038/s41598-025-94234-4] [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: 11/27/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is a chronic liver disease affecting 25% of the European population, with rising global incidence. Liver damage includes ballooning, steatosis, inflammation and fibrosis. Associated brain disorders include sleep, cognitive issues, anxiety, and depression. While neurological complications in advanced MASLD are well documented, early cerebral manifestations remain largely unexplored. This study aimed at developing an MASLD rat model to assess the onset of early brain damage, focusing on impairments of the circadian cycle rhythm and associated neuroinflammation. Sprague Dawley rats were divided into two groups: one received a high-fat, high-cholesterol (HFHC) diet for 90 days, while the other received a standard diet. Histological analysis showed significant hepatic steatosis, ballooning, and inflammation in the HFHC group (p < 0.01). These lesions correlated with elevated hepatic triglycerides (p < 0.01), increased Alanine Aminotransferase, Aspartate Aminotransferase, total cholesterol, and low-density lipoprotein, alongside decreased plasma high-density lipoprotein. Behavioural analysis using activity wheels revealed that the HFHC rats steadily maintained their activity level during the rest periods when compared with controls (p < 0.05). This behavioural alteration occurred alongside neuroinflammation, demonstrated by changes in the expression of 36 and 17 inflammatory mediators in the cerebellum and frontal cortex respectively. These changes were associated with an increase in the expression of glial cell markers (Aif1 and Gfap genes) and an increase in the number of microglial cells, affecting the frontal cortex and cerebellum differently. This rat model of early MASLD shows circadian rhythm disturbances, which could reflect sleep disorders in humans. These early brain disturbances specific to MASLD, which occur before the symptoms of liver disease become clinically apparent, could therefore be used as an early diagnosis marker for MASLD patients.
Collapse
Affiliation(s)
- Paul-Henri Graindorge
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Justine Paoli
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Berivan Yildirim
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Chloe Morel
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Ameziane Herzine
- Plateforme animalerie - Orleans University, 1 Rue de Chartes, 45067, Orléans, France
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Maud Collin
- Animalerie Campus Brabois Santé - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Isabelle Gallais
- IRSET, INSERM UMR_S 1085, Rennes University & EHESP, 2 Avenue du Professeur Léon Bernard, 35000, Rennes, France
| | - Stephane Boucard
- Dept Toxicologie et Biométrologie, Institut National de Recherche et de Sécurité pour la prevention des accidents du travail et des maladies professionnelles (INRS), 1 Rue du Morvan, 54519, Vandœuvre-Lès-Nancy, France
| | - Benoît Pouyatos
- Dept Toxicologie et Biométrologie, Institut National de Recherche et de Sécurité pour la prevention des accidents du travail et des maladies professionnelles (INRS), 1 Rue du Morvan, 54519, Vandœuvre-Lès-Nancy, France
- DevAH, UR3450, Université de Lorraine-Campus Brabois-Santé, 54519, Vandœuvre-Lès-Nancy, France
| | - David Meyre
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Dominique Lagadic-Gossmann
- IRSET, INSERM UMR_S 1085, Rennes University & EHESP, 2 Avenue du Professeur Léon Bernard, 35000, Rennes, France
| | - Odile Sergent
- IRSET, INSERM UMR_S 1085, Rennes University & EHESP, 2 Avenue du Professeur Léon Bernard, 35000, Rennes, France
| | - Henri Schroeder
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France
| | - Nathalie Grova
- UMR Inserm 1256 nGERE - Lorraine University, 9 Avenue de La Forêt de Haye, 54500, Vandœuvre-Lès-Nancy, France.
- UMR Inserm 1256 NGERE University of Lorraine, B.P. 184, 54511, Vandœuvre-Lès-Nancy, Nancy, France.
| |
Collapse
|
2
|
Subramani M, Lambrecht B, Ahmad I. Human microglia-derived proinflammatory cytokines facilitate human retinal ganglion cell development and regeneration. Stem Cell Reports 2024; 19:1092-1106. [PMID: 39059376 PMCID: PMC11368696 DOI: 10.1016/j.stemcr.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Microglia (μG), the resident immune cells in the central nervous system, surveil the parenchyma to maintain the structural and functional homeostasis of neurons. Besides, they influence neurogenesis and synaptogenesis through complement-mediated phagocytosis. Emerging evidence suggests that μG may also influence development through proinflammatory cytokines. Here, we examined the premise that tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β), the two most prominent components of the μG secretome, influence retinal development, specifically the morphological and functional differentiation of human retinal ganglion cells (hRGCs). Using controlled generation of hRGCs and human μG (hμG) from pluripotent stem cells, we demonstrate that TNF-α and IL-1β secreted by unchallenged hμG did not influence hRGC generation. However, their presence significantly facilitated neuritogenesis along with the basal function of hRGCs, which involved the recruitment of the AKT/mTOR pathway. We present ex vivo evidence that proinflammatory cytokines may play an important role in the morphological and physiological maturation of hRGCs, which may be recapitulated for regeneration.
Collapse
Affiliation(s)
- Murali Subramani
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brandon Lambrecht
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Iqbal Ahmad
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
3
|
Tang J, Zhao S, Shi H, Li X, Ran L, Cao J, He Y. Effects on peripheral and central nervous system of key inflammatory intercellular signalling peptides and proteins in psoriasis. Exp Dermatol 2024; 33:e15104. [PMID: 38794817 DOI: 10.1111/exd.15104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
Psoriasis is a chronic systemic inflammatory cutaneous disease. Where the immune system plays an important role in its pathogenesis, with key inflammatory intercellular signalling peptides and proteins including IL-17 and IL-23. The psychoneurological system also figures prominently in development of psoriasis. There is a high prevalence of comorbidity between psoriasis and mental health disorders such as depression, anxiety and mania. Patients with psoriasis often suffer from pathological pain in the lesions, and their neurological accidents could improve the lesions in innervated areas. The immune system and the psychoneurological system interact closely in the pathogenesis of psoriasis. Patients with psoriasis exhibit abnormal levels of neuropeptides both in circulating and localized lesion, acting as immunomodulators involved in the inflammatory response. Moreover, receptors for inflammatory factors are expressed in both peripheral and central nervous systems (CNSs), suggesting that nervous system can receive and be influenced by signals from immune system. Key inflammatory intercellular signalling peptides and proteins in psoriasis, such as IL-17 and IL-23, can be involved in sensory signalling and may affect synaptic plasticity and the blood-brain barrier of CNS through the circulation. This review provides an overview of the multiple effects on the peripheral and CNS under conditions of systemic inflammation in psoriasis, providing a framework and inspiration for in-depth studies of neuroimmunomodulation in psoriasis.
Collapse
Affiliation(s)
- Jue Tang
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Siqi Zhao
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huijuan Shi
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuan Li
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Liwei Ran
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiali Cao
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yanling He
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Branch in Beijing Chaoyang Hospital, Beijing, China
| |
Collapse
|
4
|
Halepoto DM, AL-Ayadhi LY, Alhowikan AM, Elamin NE. Role of autoimmunity in Neuronal damage in children with Autism spectrum disorder. Pak J Med Sci 2023; 39:1858-1864. [PMID: 37936741 PMCID: PMC10626070 DOI: 10.12669/pjms.39.6.6804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 04/10/2023] [Accepted: 04/24/2023] [Indexed: 11/09/2023] Open
Abstract
"Autism spectrum disorder (ASD) is complex neurodevelopmental disorder characterized by impairments in three core behavioral: social deficits, impaired communication, and repetitive behaviors." There is developing indication and emerging data that irregular autoimmune responses to the central nervous system may play a pathogenic role in patients with autism spectrum disorder." The aim of this review was to discuss the updated research carried out at Autism research and treatment center, King Saud University, Riyadh, Kingdom of Saudi Arabia particularly on the role of autoimmunity in Autism spectrum disorder. This review also present state of information available about the role of autoimmunity biomarkers involved in the neuronal damage of central nervous system in autistic children. The systematic literature search was carried out using Google Scholar, Science direct and PubMed databases on the role of autoimmunity in autism and reviewed all relevant articles published in peer reviewed journals by Autism research and treatment center, King Saud University, Riyadh, Kingdom of Saudi Arabia till April, 2022. We searched relevant articles using key words Autism spectrum disorder, Autoimmunity, Neuroinflamation and Central nervous system. This review revealed that plasma levels of autoimmunity related factors/ markers were altered in patients with autism. Significant change in blood markers in subjects with ASD may resulted in several years of decreased neutrotrophic support along with increasing impairment in relationship with down-regulated inflammation that may play a role in the ASD. Overall, the role of autoimmunity in ASD subjects with excess of anti-brain antibodies suggest that in some patients, autoantibodies that target the CNS may be pathological factor in neuronal growth in autistic children. Large cohort studies with well-defined and specially pheno typed autistic groups and matched healthy controls are required to examine the role of autoantibodies in the pathology of subjects with ASD.
Collapse
Affiliation(s)
- Dost Muhammad Halepoto
- Dr. Dost Muhammad Halepoto, PhD. Autism Research and Treatment center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Laila Yousif AL-Ayadhi
- Dr. Laila Yousif AL-Ayadhi, PhD. Autism Research and Treatment center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Mohammed Alhowikan
- Dr. Abdulrahman Mohammed Alhowikan, PhD. Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Nadra Elyass Elamin
- Dr. Nadra Elyass Elamin, PhD. Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Lima TS. Beyond an inflammatory mediator: Interleukin-1 in neurophysiology. Exp Physiol 2023; 108:917-924. [PMID: 37031383 PMCID: PMC10988528 DOI: 10.1113/ep090780] [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: 01/18/2023] [Accepted: 03/20/2023] [Indexed: 04/10/2023]
Abstract
NEW FINDINGS What is the topic of this review? This review focuses on the physiological role of the cytokine interleukin-1β in the CNS. What advances does it highlight? Traditionally, interleukin-1β is known as a key mediator of inflammation and immunity. This review highlights the more recent findings describing how interleukin-1β signalling is required to maintain homeostasis in the CNS. ABSTRACT Since its discovery in the early 1940s, the interleukin-1 (IL-1) cytokine family has been associated primarily with acute and chronic inflammation. The family member IL-1β is produced by different leucocytes, endothelial cells and epithelial cells. This cytokine has been characterized as a key modulator of inflammation and innate immunity because it induces the transcription of several downstream inflammatory genes. More recently, several groups have demonstrated that IL-1β production is also required to maintain homeostasis in several organ systems. This review focuses on providing an overview of the more recently characterized role of IL-1β in the physiology of the CNS. So far, IL-1β signalling has been implicated in neuronal survival, neurite growth, synaptic pruning, synaptic transmission, neuroplasticity and neuroendocrine functions.
Collapse
Affiliation(s)
- Tatiane S. Lima
- Department of Biological SciencesCalifornia State Polytechnic UniversityPomonaCaliforniaUSA
| |
Collapse
|
6
|
Kropp KA, Sun G, Viejo-Borbolla A. Colonization of peripheral ganglia by herpes simplex virus type 1 and 2. Curr Opin Virol 2023; 60:101333. [PMID: 37267706 DOI: 10.1016/j.coviro.2023.101333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/10/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
Herpes simplex virus type 1 (HSV-1) and 2 (HSV-2) infect and establish latency in neurons of the peripheral nervous system to persist lifelong in the host and to cause recurrent disease. During primary infection, HSV replicates in epithelial cells in the mucosa and skin and then infects neurites, highly dynamic structures that grow or retract in the presence of attracting or repelling cues, respectively. Following retrograde transport in neurites, HSV establishes latency in the neuronal nucleus. Viral and cellular proteins participate in the chromatinization of the HSV genome that regulates gene expression, persistence, and reactivation. HSV-2 modulates neurite outgrowth during primary infection and upon reactivation, probably to facilitate infection and survival of neurons. Whether HSV-1 modulates neurite outgrowth and the underlying mechanism is currently under investigation. This review deals with HSV-1 and HSV-2 colonization of peripheral neurons, with a focus on the modulation of neurite outgrowth by these viruses.
Collapse
Affiliation(s)
- Kai A Kropp
- Institute of Virology, Hannover Medical School, Carl-Neuberg Strasse 1, Hannover, Germany
| | - Guorong Sun
- Institute of Virology, Hannover Medical School, Carl-Neuberg Strasse 1, Hannover, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Carl-Neuberg Strasse 1, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
7
|
Liang MZ, Lu TH, Chen L. Timely expression of PGAM5 and its cleavage control mitochondrial homeostasis during neurite re-growth after traumatic brain injury. Cell Biosci 2023; 13:96. [PMID: 37221611 DOI: 10.1186/s13578-023-01052-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/13/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Patients suffered from severe traumatic brain injury (TBI) have twice the risk of developing into neurodegenerative diseases later in their life. Thus, early intervention is needed not only to treat TBI but also to reduce neurodegenerative diseases in the future. Physiological functions of neurons highly depend on mitochondria. Thus, when mitochondrial integrity is compromised by injury, neurons would initiate a cascade of events to maintain homeostasis of mitochondria. However, what protein senses mitochondrial dysfunction and how mitochondrial homeostasis is maintained during regeneration remains unclear. RESULTS We found that TBI-increased transcription of a mitochondrial protein, phosphoglycerate mutase 5 (PGAM5), during acute phase was via topological remodeling of a novel enhancer-promoter interaction. This up-regulated PGAM5 correlated with mitophagy, whereas presenilins-associated rhomboid-like protein (PARL)-dependent PGAM5 cleavage at a later stage of TBI enhanced mitochondrial transcription factor A (TFAM) expression and mitochondrial mass. To test whether PGAM5 cleavage and TFAM expression were sufficient for functional recovery, mitochondrial oxidative phosphorylation uncoupler carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP) was used to uncouple electron transport chain and reduce mitochondrial function. As a result, FCCP triggered PGAM5 cleavage, TFAM expression and recovery of motor function deficits of CCI mice. CONCLUSIONS Findings from this study implicate that PGAM5 may act as a mitochondrial sensor for brain injury to activate its own transcription at acute phase, serving to remove damaged mitochondria through mitophagy. Subsequently, PGAM5 is cleaved by PARL, and TFAM expression is increased for mitochondrial biogenesis at a later stage after TBI. Taken together, this study concludes that timely regulation of PGAM5 expression and its own cleavage are required for neurite re-growth and functional recovery.
Collapse
Affiliation(s)
- Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Ting-Hsuan Lu
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
8
|
Manjili MH. The adaptation model of immunity: A new insight into aetiology and treatment of multiple sclerosis. Scand J Immunol 2023; 97:e13255. [PMID: 36680379 DOI: 10.1111/sji.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/04/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Current research and drug development for multiple sclerosis (MS) is fully influenced by the self-nonself (SNS) model of immunity, suggesting that breakage of immunological tolerance towards self-antigens expressed in the central nervous system (CNS) is responsible for pathogenesis of MS; thus, immune suppressive drugs are recommended for the management of the disease. However, this model provides incomplete understanding of the causes and pathways involved in the onset and progression of MS and limits our ability to effectively treat this neurological disease. Some pre-clinical and clinical reports have been misunderstood; some others have been underappreciated because of the lack of a theoretical model that can explain them. Also, current immunotherapies are guided according to the models that are not designed to explain the functional outcomes of an immune response. The adaptation model of immunity is proposed to offer a new understanding of the existing data and galvanize a new direction for the treatment of MS. According to this model, the immune system continuously communicates with the CNS through the adaptation receptors (AdRs) and adaptation ligands (AdLs) or co-receptors, signal IV, to support cell growth and neuroplasticity. Alterations in the expression of the neuronal AdRs results in MS by shifting the cerebral inflammatory immune responses from remyelination to demyelination. Therefore, novel therapeutics for MS should be focused on the discovery and targeting of the AdR/AdL axis in the CNS rather than carrying on with immune suppressive interventions.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, Virginia, USA
| |
Collapse
|
9
|
Jo D, Jung YS, Song J. Lipocalin-2 Secreted by the Liver Regulates Neuronal Cell Function Through AKT-Dependent Signaling in Hepatic Encephalopathy Mouse Model. Clin Nutr Res 2023; 12:154-167. [PMID: 37214781 PMCID: PMC10193436 DOI: 10.7762/cnr.2023.12.2.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 05/24/2023] Open
Abstract
Hepatic encephalopathy (HE) associated with liver failure is accompanied by hyperammonemia, severe inflammation, depression, anxiety, and memory deficits as well as liver injury. Recent studies have focused on the liver-brain-inflammation axis to identify a therapeutic solution for patients with HE. Lipocalin-2 is an inflammation-related glycoprotein that is secreted by various organs and is involved in cellular mechanisms including iron homeostasis, glucose metabolism, cell death, neurite outgrowth, and neurogenesis. In this study, we investigated that the roles of lipocalin-2 both in the brain cortex of mice with HE and in Neuro-2a (N2A) cells. We detected elevated levels of lipocalin-2 both in the plasma and liver in a bile duct ligation mouse model of HE. We confirmed changes in cytokine expression, such as interleukin-1β, cyclooxygenase 2 expression, and iron metabolism related to gene expression through AKT-mediated signaling both in the brain cortex of mice with HE and N2A cells. Our data showed negative effects of hepatic lipocalin-2 on cell survival, iron homeostasis, and neurite outgrowth in N2A cells. Thus, we suggest that regulation of lipocalin-2 in the brain in HE may be a critical therapeutic approach to alleviate neuropathological problems focused on the liver-brain axis.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| | - Yoon Seok Jung
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| |
Collapse
|
10
|
Shichita T, Ooboshi H, Yoshimura A. Neuroimmune mechanisms and therapies mediating post-ischaemic brain injury and repair. Nat Rev Neurosci 2023; 24:299-312. [PMID: 36973481 DOI: 10.1038/s41583-023-00690-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The nervous and immune systems control whole-body homeostasis and respond to various types of tissue injury, including stroke, in a coordinated manner. Cerebral ischaemia and subsequent neuronal cell death activate resident or infiltrating immune cells, which trigger neuroinflammation that affects functional prognosis after stroke. Inflammatory immune cells exacerbate ischaemic neuronal injury after the onset of brain ischaemia; however, some of the immune cells thereafter change their function to neural repair. The recovery processes after ischaemic brain injury require additional and close interactions between the nervous and immune systems through various mechanisms. Thus, the brain controls its own inflammation and repair processes after injury via the immune system, which provides a promising therapeutic opportunity for stroke recovery.
Collapse
Affiliation(s)
- Takashi Shichita
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
- Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
- Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.
| | - Hiroaki Ooboshi
- Section of Internal Medicine, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
11
|
Cao JF, Hu X, Xiong L, Wu M, Yang X, Wang C, Chen S, Xu H, Chen H, Ma X, Mi Y, Zhang X. Interference of Interleukin-1 β Mediated by Lentivirus Promotes Functional Recovery of Spinal Cord Contusion Injury in Rats via the PI3K/AKT1 Signaling Pathway. Mediators Inflamm 2022; 2022:6285099. [PMID: 39262872 PMCID: PMC11390212 DOI: 10.1155/2022/6285099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/30/2022] [Indexed: 09/13/2024] Open
Abstract
Purpose Inflammation and apoptosis after spinal cord contusion (SCC) are important causes of irreversible spinal cord injury. Interleukin-1β (IL-1β) is a key inflammatory factor that promotes the aggravation of spinal cord contusion. However, the specific role and regulatory mechanism of IL-1β in spinal cord contusion is still unclear. Therefore, this study applied bioinformatics to analyze and mine potential gene targets interlinked with IL-1β, animal experiments and lentiviral interference technology were used to explore whether IL-1β affected the recovery of motor function in spinal cord contusion by interfering with PI3K/AKT1 signaling pathway. Method This study used bioinformatics to screen and analyze gene targets related to IL-1β. The rat SCC animal model was established by the Allen method, and the Basso Beattie Bresnahan (BBB) score was used to evaluate the motor function of the spinal cord-injured rats. Immunohistochemistry and immunofluorescence were used to localize the expression of IL-1β and AKT1 proteins in spinal cord tissue. Quantitative polymerase chain reaction and Western blot were used to detect the gene and protein expressions of IL-1β, PI3K, and AKT1. RNAi technology was used to construct lentivirus to inhibit the expression of IL-1β, lentiviral interference with IL-1β was used to investigate the effect of IL-1β and AKT1 on the function of spinal cord contusion and the relationship among IL-1β, AKT1, and downstream signaling pathways. Results Bioinformatics analysis suggested a close relationship between IL-1β and AKT1. Animal experiments have confirmed that IL-1β is closely related to the functional recovery of spinal cord contusion. Firstly, from the phenomenological level, the BBB score decreased after SCC, IL-1β and AKT1 were located in the cytoplasm of neurons in the anterior horn of the spinal cord, and the expression levels of IL-1β gene and protein in the experimental group were higher than those in the sham operation group. At the same time, the expression of AKT1 gene decreased, the results suggested that the increase of IL-1β affected the functional recovery of spinal cord contusion. Secondly, from the functional level, after inhibiting the expression of IL-1β with a lentivirus-mediated method, the BBB score was significantly increased, and the motor function of the spinal cord was improved. Thirdly, from the mechanistic level, bioinformatics analysis revealed the relationship between IL-1β and AKT1. In addition, the experiment further verified that in the PI3K/AKT1 signaling pathway, inhibition of IL-1β expression upregulated AKT1 gene expression, but PI3K expression was unchanged. Conclusion Inhibition of IL-1β promotes recovery of motor function after spinal cord injury in rats through upregulation of AKT1 expression in the PI3K/AKT1 signaling pathway. Bioinformatics analysis suggested that IL-1β may affect apoptosis and regeneration by inhibiting the expression of AKT1 in the PI3K/AKT1 signaling pathway to regulate the downstream FOXO, mTOR, and GSK3 signaling pathways; thereby hindering the recovery of motor function in rats after spinal cord contusion. It provided a new perspective for clinical treatment of spinal cord contusion in the future.
Collapse
Affiliation(s)
- Jun-Feng Cao
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xi Hu
- Taikang Tongji Wuhan Hospital, Wuhan, China
| | - Li Xiong
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Mei Wu
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Xingyu Yang
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Chaochao Wang
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Shengyan Chen
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Hengxiang Xu
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Huanyu Chen
- Basic Medical College of Chengdu Medical College, Chengdu, China
| | - Xuntai Ma
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yongjie Mi
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- National Demonstration Center for Experimental Clinical Medicine Education of Chengdu Medical College, Chengdu, China
| | - Xiao Zhang
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- Basic Medical College of Chengdu Medical College, Chengdu, China
- National Demonstration Center for Experimental Clinical Medicine Education of Chengdu Medical College, Chengdu, China
| |
Collapse
|
12
|
Shoji M, Okamoto R, Unno T, Harada K, Kubo M, Fukuyama Y, Kuzuhara T. Transcriptome analysis of PC12 cells reveals that trans-banglene upregulates RT1-CE1 and downregulates abca1 in the neurotrophic pathway. Biol Pharm Bull 2022; 45:1784-1790. [PMID: 36155550 DOI: 10.1248/bpb.b22-00474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Trans(t)-banglene and cis(c)-banglene possess neurotrophin-like activity in rat neurons. However, the molecular mechanisms underlying t-banglene-induced neurotrophic activity in rat and human neurons remain unclear. Here, we performed transcriptome analysis in PC12 cells, a rat adrenal gland pheochromocytoma cell line treated with t-banglene, using comprehensive RNA sequencing. The differentially expressed gene analysis of the sequencing data revealed that the expression of RT1 class I, locus CE1 (RT1-CE1) was upregulated, and that of ATP binding cassette subfamily A member 1 (abca1), myosin light chain 6, and hippocampus abundant transcript 1 was downregulated in t-banglene-treated PC12 cells, with statistically significant differences. We also confirmed the RT1-CE1 upregulation and abca1 downregulation in t-banglene-treated PC12 cells by reverse transcription quantitative real-time polymerase chain reaction. RT1-CEl is a major histocompatibility complex class I (MHCI) protein. ABCAl is a major cholesterol transporter that regulates efflux of intracellular cholesterol and phospholipids. Thus, our results suggest an exciting link between MHCI, cholesterol regulation, and neural development.
Collapse
Affiliation(s)
- Masaki Shoji
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Risa Okamoto
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Taishi Unno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Kenichi Harada
- Laboratory of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Miwa Kubo
- Laboratory of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Yoshiyasu Fukuyama
- Laboratory of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Takashi Kuzuhara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
13
|
Cui Y, Zhang NN, Wang D, Meng WH, Chen HS. Modified Citrus Pectin Alleviates Cerebral Ischemia/Reperfusion Injury by Inhibiting NLRP3 Inflammasome Activation via TLR4/NF-ĸB Signaling Pathway in Microglia. J Inflamm Res 2022; 15:3369-3385. [PMID: 35706530 PMCID: PMC9191615 DOI: 10.2147/jir.s366927] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/28/2022] [Indexed: 12/19/2022] Open
Abstract
Background Galectin-3 acts as a mediator of microglial inflammatory response following stroke injury. However, it remains unclear whether inhibiting galectin-3 protects against cerebral ischemia/reperfusion injury. We aimed to investigate the neuroprotective effects of modified citrus pectin (MCP, a galectin-3 blocker) in ischemic stroke and underlying mechanisms. Methods The middle cerebral artery occlusion/reperfusion (MCAO/R) model in C57BL/6J mice and oxygen-glucose deprivation/reoxygenation (ODG/R) model in neuronal (HT-22) and microglial (BV-2) cells were utilized in the following experiments: 1) the neuroprotective effects of MCP with different concentrations were evaluated in vivo and in vitro through measuring neurological deficit scores, brain water content, infarction volume, cell viability, and cell apoptosis; 2) the mechanisms of its neuroprotection were explored in mice and microglial cells through detecting the expression of NLRP3 (NOD-like receptor 3) inflammasome-related proteins by immunofluorescence staining and Western blotting analyses. Results Among the tested concentrations, 800 mg/kg/d MCP in mice and 4 g/L MCP in cells, respectively, showed in vivo and in vitro neuroprotective effects on all the tests, compared with vehicle group. First, MCP significantly reduced neurological deficit scores, brain water content and infarction volume, and alleviated cell injury in the cerebral cortex of MCAO/R model. Second, MCP increased cell viability and reduced cell apoptosis in the neuronal OGD/R model. Third, MCP blocked galectin-3 and decreased the expression of TLR4 (Toll-like receptor 4)/NF-κBp65 (nuclear factor kappa-B)/NLRP3/cleaved-caspase-1/IL-1β (interleukin-1β) in microglial cells. Conclusion This is the first report that MCP exerts neuroprotective effects in ischemic stroke through blocking galectin-3, which may be mediated by inhibiting the activation of NLRP3 inflammasome via TLR4/NF-κB signaling pathway in microglia.
Collapse
Affiliation(s)
- Yu Cui
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, People's Republic of China.,Department of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Nan-Nan Zhang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Dan Wang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| | - Wei-Hong Meng
- Department of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, People's Republic of China
| |
Collapse
|
14
|
GIT1 Promotes Axonal Growth in an Inflammatory Environment by Promoting the Phosphorylation of MAP1B. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7474177. [PMID: 35340202 PMCID: PMC8942666 DOI: 10.1155/2022/7474177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/11/2022] [Accepted: 01/25/2022] [Indexed: 11/21/2022]
Abstract
Spinal cord injury (SCI) is a severe traumatic condition. The loss of the bundle of axons involved in motor conduction in the spinal cord after SCI is the main cause of motor function injury. Presently, axon regeneration in the spinal cord has been studied extensively, but it remains unclear how axon growth is regulated in an inflammatory environment at the cellular level. In the present study, GIT1 knockout (KO) mouse neurons were cultured in a microfluidic device to simulate the growth of axons in an inflammatory environment. The molecular regulation of axon growth in an inflammatory environment by GIT1 was then investigated. We found that the axon growth of GIT1 KO mouse neurons was restricted in an inflammatory environment. Further investigations revealed that in both axons and cell bodies in the inflammatory environment, GIT1 phosphorylated ERK, promoted the entry of Nrf2 into the nucleus, and promoted the transcription of MAP1B, thereby increasing the levels of MAP1B and p-MAP1B and promoting axon growth. We also found that MAP1B could be translated locally in axons and transported in cell bodies and axons. In conclusion, we found that GIT1 regulated axon growth in an inflammatory environment. This provided a theoretical basis for axon regeneration in an inflammatory environment after SCI to develop new treatment options for axon regeneration.
Collapse
|
15
|
Solek CM, Farooqi NAI, Brake N, Kesner P, Schohl A, Antel JP, Ruthazer ES. Early Inflammation Dysregulates Neuronal Circuit Formation In Vivo via Upregulation of IL-1β. J Neurosci 2021; 41:6353-6366. [PMID: 34103360 PMCID: PMC8287996 DOI: 10.1523/jneurosci.2159-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 04/04/2021] [Accepted: 05/29/2021] [Indexed: 12/18/2022] Open
Abstract
Neuroimmune interaction during development is strongly implicated in the pathogenesis of neurodevelopmental disorders, but the mechanisms that cause neuronal circuit dysregulation are not well understood. We performed in vivo imaging of the developing retinotectal system in the larval zebrafish to characterize the effects of immune system activation on refinement of an archetypal sensory processing circuit. Acute inflammatory insult induced hyperdynamic remodeling of developing retinal axons in larval fish and increased axon arbor elaboration over days. Using calcium imaging in GCaMP6s transgenic fish, we showed that these morphologic changes were accompanied by a shift toward decreased visual acuity in tectal cells. This finding was supported by poorer performance in a visually guided behavioral task. We further found that the pro-inflammatory cytokine, interleukin-1β (IL-1β), is upregulated by the inflammatory insult, and that downregulation of IL-1β abrogated the effects of inflammation on axonal dynamics and growth. Moreover, baseline branching of the retinal ganglion cell arbors in IL-1β morphant animals was significantly different from that in control larvae, and their performance in a predation assay was impaired, indicating a role for this cytokine in normal neuronal development. This work establishes a simple and powerful non-mammalian model of developmental immune activation and demonstrates a role for IL-1β in mediating the pathologic effects of inflammation on neuronal circuit development.SIGNIFICANCE STATEMENT Maternal immune activation can increase the risk of neurodevelopmental disorders in offspring; however, the mechanisms involved are not fully understood. Using a non-mammalian vertebrate model of developmental immune activation, we show that even brief activation of inflammatory pathways has immediate and long-term effects on the arborization of axons, and that these morphologic changes have functional and behavioral consequences. Finally, we show that the pro-inflammatory cytokine IL-1β plays an essential role in both the effects of inflammation on circuit formation and normal axonal development. Our data add to a growing body of evidence supporting epidemiological studies linking immune activation to neurodevelopmental disorders, and help shed light on the molecular and cellular processes that contribute to the etiology of these disorders.
Collapse
Affiliation(s)
- Cynthia M Solek
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Nasr A I Farooqi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Niklas Brake
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Philip Kesner
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Anne Schohl
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack P Antel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Edward S Ruthazer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
16
|
Zengeler KE, Lukens JR. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat Rev Immunol 2021; 21:454-468. [PMID: 33479477 PMCID: PMC9213174 DOI: 10.1038/s41577-020-00487-7] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/29/2022]
Abstract
The immune and nervous systems have unique developmental trajectories that individually build intricate networks of cells with highly specialized functions. These two systems have extensive mechanistic overlap and frequently coordinate to accomplish the proper growth and maturation of an organism. Brain resident innate immune cells - microglia - have the capacity to sculpt neural circuitry and coordinate copious and diverse neurodevelopmental processes. Moreover, many immune cells and immune-related signalling molecules are found in the developing nervous system and contribute to healthy neurodevelopment. In particular, many components of the innate immune system, including Toll-like receptors, cytokines, inflammasomes and phagocytic signals, are critical contributors to healthy brain development. Accordingly, dysfunction in innate immune signalling pathways has been functionally linked to many neurodevelopmental disorders, including autism and schizophrenia. This review discusses the essential roles of microglia and innate immune signalling in the assembly and maintenance of a properly functioning nervous system.
Collapse
Affiliation(s)
- Kristine E Zengeler
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), Charlottesville, VA, USA.
- Neuroscience Graduate Program, Charlottesville, VA, USA.
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - John R Lukens
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), Charlottesville, VA, USA.
- Neuroscience Graduate Program, Charlottesville, VA, USA.
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
17
|
Neuroinflammation: An Integrating Overview of Reactive-Neuroimmune Cell Interactions in Health and Disease. Mediators Inflamm 2021; 2021:9999146. [PMID: 34158806 PMCID: PMC8187052 DOI: 10.1155/2021/9999146] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
The concept of central nervous system (CNS) inflammation has evolved over the last decades. Neuroinflammation is the response of reactive CNS components to altered homeostasis, regardless of the cause to be endogenous or exogenous. Neurological diseases, whether traumatic, neoplastic, ischemic, metabolic, toxic, infectious, autoimmune, developmental, or degenerative, involve direct and indirect immune-related neuroinflammation. Brain infiltrates of the innate and adaptive immune system cells appear in response to an infective or otherwise noxious agent and produce inflammatory mediators. Mediators of inflammation include local and recruited cells and signals. Processes derived from extrinsic and intrinsic CNS diseases also elicit the CNS inflammatory response. A deeper understanding of immune-related inflammation in health and disease is necessary to find potential therapeutic targets for preventing or reducing CNS damage. This review is aimed at discussing the innate and adaptive immune system functions and their roles in regulating brain cell responses in disease and homeostasis maintenance.
Collapse
|
18
|
Smith DR, Dumont CM, Park J, Ciciriello AJ, Guo A, Tatineni R, Cummings BJ, Anderson AJ, Shea LD. Polycistronic Delivery of IL-10 and NT-3 Promotes Oligodendrocyte Myelination and Functional Recovery in a Mouse Spinal Cord Injury Model. Tissue Eng Part A 2020; 26:672-682. [PMID: 32000627 DOI: 10.1089/ten.tea.2019.0321] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
One million estimated cases of spinal cord injury (SCI) have been reported in the United States and repairing an injury has constituted a difficult clinical challenge. The complex, dynamic, inhibitory microenvironment postinjury, which is characterized by proinflammatory signaling from invading leukocytes and lack of sufficient factors that promote axonal survival and elongation, limits regeneration. Herein, we investigated the delivery of polycistronic vectors, which have the potential to coexpress factors that target distinct barriers to regeneration, from a multiple channel poly(lactide-co-glycolide) (PLG) bridge to enhance spinal cord regeneration. In this study, we investigated polycistronic delivery of IL-10 that targets proinflammatory signaling, and NT-3 that targets axonal survival and elongation. A significant increase was observed in the density of regenerative macrophages for IL-10+NT-3 condition relative to conditions without IL-10. Furthermore, combined delivery of IL-10+NT-3 produced a significant increase of axonal density and notably myelinated axons compared with all other conditions. A significant increase in functional recovery was observed for IL-10+NT-3 delivery at 12 weeks postinjury that was positively correlated to oligodendrocyte myelinated axon density, suggesting oligodendrocyte-mediated myelination as an important target to improve functional recovery. These results further support the use of multiple channel PLG bridges as a growth supportive substrate and platform to deliver bioactive agents to modulate the SCI microenvironment and promote regeneration and functional recovery. Impact statement Spinal cord injury (SCI) results in a complex microenvironment that contains multiple barriers to regeneration and functional recovery. Multiple factors are necessary to address these barriers to regeneration, and polycistronic lentiviral gene therapy represents a strategy to locally express multiple factors simultaneously. A bicistronic vector encoding IL-10 and NT-3 was delivered from a poly(lactide-co-glycolide) bridge, which provides structural support that guides regeneration, resulting in increased axonal growth, myelination, and subsequent functional recovery. These results demonstrate the opportunity of targeting multiple barriers to SCI regeneration for additive effects.
Collapse
Affiliation(s)
- Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida.,Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, Miami, Florida
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky.,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Amina Guo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Ravindra Tatineni
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Brian J Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California.,Department of Anatomy and Neurobiology, University of California, Irvine, California.,Department of Physical Medicine and Rehabilitation, University of California, Irvine, California
| | - Aileen J Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California.,Department of Anatomy and Neurobiology, University of California, Irvine, California.,Department of Physical Medicine and Rehabilitation, University of California, Irvine, California
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
19
|
Lee Y, Lee S, Chang SC, Lee J. Significant roles of neuroinflammation in Parkinson's disease: therapeutic targets for PD prevention. Arch Pharm Res 2019; 42:416-425. [PMID: 30830660 DOI: 10.1007/s12272-019-01133-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023]
Abstract
Glial cells outnumber neurons in the brain and play important roles in the neuroinflammation that accompanies brain damage in neurodegenerative diseases. In Parkinson's disease (PD), dopaminergic neuronal loss is accompanied by inflammatory changes in microglia, astrocytes, innate immune cells, and infiltrating peripheral immune cells. Neuroinflammation is probably a fundamental immune response to protect neurons from harm and compensate for neuronal damage, but at the same time, its neurotoxic effects exacerbate neuron damage. Furthermore, neuroinflammatory response is regulated by immune cells, such as microglia, astrocytes, and peripheral immune cells, and by cytokines and chemokines. Accordingly, it is crucial that we understand how such immune cells in the brain regulate neuroinflammatory responses in PD pathology. This review describes the roles played by glia-mediated neuroinflammation in PD, both good and bad, and the therapeutic strategies used to treat PD.
Collapse
Affiliation(s)
- Yujeong Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seulah Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seung-Cheol Chang
- Institute of BioPhysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
20
|
Tsarouchas TM, Wehner D, Cavone L, Munir T, Keatinge M, Lambertus M, Underhill A, Barrett T, Kassapis E, Ogryzko N, Feng Y, van Ham TJ, Becker T, Becker CG. Dynamic control of proinflammatory cytokines Il-1β and Tnf-α by macrophages in zebrafish spinal cord regeneration. Nat Commun 2018; 9:4670. [PMID: 30405119 PMCID: PMC6220182 DOI: 10.1038/s41467-018-07036-w] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 10/12/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury leads to a massive response of innate immune cells in non-regenerating mammals, but also in successfully regenerating zebrafish. However, the role of the immune response in successful regeneration is poorly defined. Here we show that inhibiting inflammation reduces and promoting it accelerates axonal regeneration in spinal-lesioned zebrafish larvae. Mutant analyses show that peripheral macrophages, but not neutrophils or microglia, are necessary for repair. Macrophage-less irf8 mutants show prolonged inflammation with elevated levels of Tnf-α and Il-1β. Inhibiting Tnf-α does not rescue axonal growth in irf8 mutants, but impairs it in wildtype animals, indicating a pro-regenerative role of Tnf-α. In contrast, decreasing Il-1β levels or number of Il-1β+ neutrophils rescue functional regeneration in irf8 mutants. However, during early regeneration, interference with Il-1β function impairs regeneration in irf8 and wildtype animals. Hence, inflammation is dynamically controlled by macrophages to promote functional spinal cord regeneration in zebrafish. While proinflammatory signalling is preventive to axon regrowth, activated macrophages can be beneficial, for example by limiting the inflammation. This study uses mutant zebrafish lines that lack macrophages and/or microglia to show that peripheral macrophages are necessary in axon regrowth following complete transection of spinal cord.
Collapse
Affiliation(s)
- Themistoklis M Tsarouchas
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.,Technische Universität Dresden, DFG-Center of Regenerative Therapies Dresden, Fetscherstraße 105, Dresden, 01307, Germany
| | - Leonardo Cavone
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Tahimina Munir
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Marvin Lambertus
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.,Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0316, Oslo, Norway
| | - Anna Underhill
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Thomas Barrett
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Elias Kassapis
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Nikolay Ogryzko
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Yi Feng
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
21
|
Schwarz AP, Rotov AY, Chuprina OI, Krytskaya DU, Trofimov AN, Kosheverova VV, Ischenko AM, Zubareva OE. Developmental prefrontal mRNA expression of D2 dopamine receptor splice variants and working memory impairments in rats after early life Interleukin-1β elevation. Neurobiol Learn Mem 2018; 155:231-238. [PMID: 30092312 DOI: 10.1016/j.nlm.2018.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 11/29/2022]
Abstract
Long (D2L) and Short (D2S) isoforms of D2 dopamine receptor differ in their biochemical and physiological properties, which could affect functioning of prefrontal cortex. Contribution of distinct D2 dopamine receptor isoforms to cognitive dysfunctions and its developmental regulation are currently not fully elucidated. In the present study, we evaluated developmental mRNA expression of D2S/D2L dopamine receptor isoforms within the rat medial prefrontal cortex (mPFC) in the model of neurodevelopmental cognitive dysfunction. Working memory performance (Y-maze spontaneous alternations) and D2S/D2L mRNA expression in the mPFC (by qRT-PCR) were evaluated in juvenile (P27), adolescent (P42-47) and adult (P75-90) rats after chronic early life treatment with proinflammatory cytokine interleukin (IL)-1β (1 µg/kg i.p. daily P15-21). It was shown that IL-1β elevation during the 3rd week of life leads to working memory deficit originating in juvenile animals and persisting into adulthood. D2S mRNA expression was strongly downregulated during adolescence, and such downregulation was exaggerated in animals injected with IL-1β during P15-21. Early life IL-1β administrations influenced developmental changes in the D2S/D2L mRNA ratio. This measure was found to be decreased in adolescent and adult control (intact and vehicle-treated) rats compared to juvenile control, while in the case of IL-1β-treated animals, the decrease in D2S/D2L ratio was observed only in adulthood but not in adolescence compared to juvenile rats. During the adolescence, D2S mRNA expression was downregulated and D2S/D2L ratio was upregulated in the mPFC of rats treated with IL-1β during the 3rd week of life compared to controls. Based on these data we conclude that changes in the developmental expression of D2 dopamine receptor splice variants within mPFC may underlie long-lasting cognitive deficit associated with neonatal pathology.
Collapse
Affiliation(s)
- Alexander P Schwarz
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia.
| | - Alexander Yu Rotov
- Laboratory of Evolution of the Sensory Organs, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez avenue 44, 199223 St. Petersburg, Russia
| | - Olga I Chuprina
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia
| | - Darya U Krytskaya
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia
| | - Alexander N Trofimov
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia
| | - Vera V Kosheverova
- Laboratory of Intracellular Membranes Dynamics, Department of the Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky avenue 4, 194064 St. Petersburg, Russia
| | - Alexander M Ischenko
- Laboratory of Protein Biochemistry, Research Institute of Highly Pure Biopreparations, Pudozhskaya street 7, 197110 St. Petersburg, Russia
| | - Olga E Zubareva
- Laboratory of Neurobiology of the Brain Integrative Functions, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, Akademika Pavlova street 12, 197376 St. Petersburg, Russia; Laboratory of Molecular Mechanisms of Neuronal Interactions, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez avenue 44, 199223 St. Petersburg, Russia
| |
Collapse
|
22
|
Park SY, Kang MJ, Han JS. Interleukin-1 beta promotes neuronal differentiation through the Wnt5a/RhoA/JNK pathway in cortical neural precursor cells. Mol Brain 2018; 11:39. [PMID: 29973222 PMCID: PMC6033214 DOI: 10.1186/s13041-018-0383-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023] Open
Abstract
Pro-inflammatory cytokine interleukin-1 beta (IL-1β) is a key mediator of inflammation and stress in the central nervous system (CNS), and is highly expressed in the developing brain. In this study, we investigated the possible role of IL-1β in neuronal differentiation of cortical neural precursor cells (NPCs). We showed that stimulation with IL-1β increased expression levels of neurotrophin-3 (NT3) and neurogenin 1 (Ngn1) and promoted neurite outgrowth. We also found that IL-1β increased mRNA and protein levels of Wnt5a. Knockdown of Wnt5a by transfection with Wnt5a siRNA inhibited IL-1β-induced neuronal differentiation. Moreover, IL-1β-induced Wnt5a expression was regulated by nuclear factor kappa B (NF-κB) activation, which is involved in IL-1β-mediated neuronal differentiation. To examine the role of Wnt5a in neuronal differentiation of NPCs, we exogenously added Wnt5a. We found that exogenous Wnt5a promotes neuronal differentiation, and activates the RhoA/Rho-associated kinase (ROCK)/c-jun N-terminal kinase (JNK) pathway. In addition, Wnt5a-induced neuronal differentiation was blocked by RhoA siRNA, as well as by a specific Rho-kinase inhibitor (Y27632) or a SAPK/JNK inhibitor (SP600125). Furthermore, treatment with RhoA siRNA, Y27632, or SP600125 suppressed the IL-1β-induced neuronal differentiation. Therefore, these results suggest that the sequential Wnt5a/RhoA/ROCK/JNK pathway is involved in IL-1β-induced neuronal differentiation of NPCs.
Collapse
Affiliation(s)
- Shin-Young Park
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Min-Jeong Kang
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Joong-Soo Han
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
23
|
Abstract
This study was undertaken to establish a method for the culture of organotypic spinal cord slices. A long-term organotypic spinal cord slice culture was conducted from postnatal rats. Lumbar spinal cord was isolated, and meninges were removed from the spinal cord. The spinal cord was embedded in 4% agarose, and was sectioned by vibratome into slices. Then the slices were cultured on the surface of the membrane inserts, which were placed in six-well plates containing 1 ml of growth medium at 37°C in an incubator with 5% humidified carbon dioxide. The cultured organotypic spinal cord slices were examined by light microscopy and immunocytochemistry. The organotypic spinal cord slices were fully attached to the membrane inserts after 10 days in vitro. The general change in color and transparency from whitish to transparent gray appeared at the seventh and eighth day. Under the light microscope, the outgrowth of cells from the edge of the living slices arose from the second day of the culture, and arose to peak at the sixth and seventh day. The organotypic spinal cord slices were characterized as clear, semitransparent structures with bright and good refraction until the 14th day of culture. The viability of the slices was excellent as assessed by the trypan blue exclusion method at the 28th day, and they were positive for NeuN and GFAP. This culture technique, which does not require complex operation skills, might be a simple and efficient method for obtaining organotypic spinal cord slices in sufficient number, high viability, and contamination-free from postnatal rats.
Collapse
|
24
|
Liu X, Quan N. Microglia and CNS Interleukin-1: Beyond Immunological Concepts. Front Neurol 2018; 9:8. [PMID: 29410649 PMCID: PMC5787061 DOI: 10.3389/fneur.2018.00008] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022] Open
Abstract
Activation of microglia and expression of the inflammatory cytokine interleukin-1 (IL-1) in the CNS have become almost synonymous with neuroinflammation. In numerous studies, increased CNS IL-1 expression and altered microglial morphology have been used as hallmarks of CNS inflammation. A central concept of how CNS IL-1 and microglia influence functions of the nervous system was derived from the notion initially generated in the peripheral immune system: IL-1 stimulates monocyte/macrophage (the peripheral counterparts of microglia) to amplify inflammation. It is increasingly clear, however, CNS IL-1 acts on other targets in the CNS and microglia participates in many neural functions that are not related to immunological activities. Further, CNS exhibits immunological privilege (although not as absolute as previously thought), rendering amplification of inflammation within CNS under stringent control. This review will analyze current literature to evaluate the contribution of immunological and non-immunological aspects of microglia/IL-1 interaction in the CNS to gain insights for how these aspects might affect health and disease in the nervous tissue.
Collapse
Affiliation(s)
- Xiaoyu Liu
- College of Medicine, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Ning Quan
- College of Medicine, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
25
|
Saghazadeh A, Ferrari CC, Rezaei N. Deciphering variability in the role of interleukin-1β in Parkinson's disease. Rev Neurosci 2018; 27:635-50. [PMID: 27166719 DOI: 10.1515/revneuro-2015-0059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 04/01/2016] [Indexed: 12/16/2022]
Abstract
Although the role of inflammation in neurodegeneration has been well acknowledged, less is known on the issue of each cytokine in specific neurodegenerative diseases. In this review, we will present evidence elucidating that interleukin-1β (IL-1β) has a multi-faceted character in pathogenesis of Parkinson's disease, which is a progressive neurodegenerative disorder. Increased levels of IL-1β were found in PD patients. Besides, PD symptoms were observed in IL-1β wild-type, but not deficient, animals. These lines of evidence suggest that IL-1β may contribute to the initiation or progression of PD. On the other hand, some studies reported decreased levels of IL-1β in PD patients. Also, genetic studies provided evidence suggesting that IL-1β may protect individuals against PD. Presumably, the broad range of IL-1β role is due to its interaction with both upstream and downstream mediators. Differences in IL-1β levels could be because of glia population (i.e. microglia and astrocytes), mitogen-activated protein kinase and nuclear factor κ light-chain-enhancer of activated B cells signaling pathways, and several mediators (including cyclooxygenase, neurotrophic factors, reactive oxygen species, caspases, heme oxygenase-1, and matrix metalloproteinases). Although far from practice at this point, unraveling theoretical therapeutic targets based on the up-down IL-1β neuroweb could facilitate the development of strategies that are likely to be used for pharmaceutical designs of anti-neurodegenerative drugs of the future.
Collapse
|
26
|
Simon DW, McGeachy M, Bayır H, Clark RS, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol 2017; 13:171-191. [PMID: 28186177 PMCID: PMC5675525 DOI: 10.1038/nrneurol.2017.13] [Citation(s) in RCA: 671] [Impact Index Per Article: 83.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The 'silent epidemic' of traumatic brain injury (TBI) has been placed in the spotlight as a result of clinical investigations and popular press coverage of athletes and veterans with single or repetitive head injuries. Neuroinflammation can cause acute secondary injury after TBI, and has been linked to chronic neurodegenerative diseases; however, anti-inflammatory agents have failed to improve TBI outcomes in clinical trials. In this Review, we therefore propose a new framework of targeted immunomodulation after TBI for future exploration. Our framework incorporates factors such as the time from injury, mechanism of injury, and secondary insults in considering potential treatment options. Structuring our discussion around the dynamics of the immune response to TBI - from initial triggers to chronic neuroinflammation - we consider the ability of soluble and cellular inflammatory mediators to promote repair and regeneration versus secondary injury and neurodegeneration. We summarize both animal model and human studies, with clinical data explicitly defined throughout this Review. Recent advances in neuroimmunology and TBI-responsive neuroinflammation are incorporated, including concepts of inflammasomes, mechanisms of microglial polarization, and glymphatic clearance. Moreover, we highlight findings that could offer novel therapeutic targets for translational and clinical research, assimilate evidence from other brain injury models, and identify outstanding questions in the field.
Collapse
Affiliation(s)
- Dennis W. Simon
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mandy McGeachy
- Department of Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Hülya Bayır
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Robert S.B. Clark
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Anesthesiology, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MA 21201, USA
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Anesthesiology, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
27
|
Wu PJ, Liu HY, Huang TN, Hsueh YP. AIM 2 inflammasomes regulate neuronal morphology and influence anxiety and memory in mice. Sci Rep 2016; 6:32405. [PMID: 27561456 PMCID: PMC5000013 DOI: 10.1038/srep32405] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
Inflammasomes are the protein assemblies that consist of inflammasome sensors, adaptor apoptosis-associated speck-like proteins containing a CARD (ASC) and inflammasome caspase. Inflammasomes sense multiple danger signals via various inflammasome sensors and consequently use caspase to trigger proteolytic processing and secretion of IL-1β cytokines. Recent studies have suggested that neurons use their own innate immune system to detect danger signals and regulate neuronal morphology. Here, we investigate whether inflammasomes, the critical components of innate immunity, participate in regulation of neuronal morphology and function. Among various sensors, Absent in melanoma 2 (Aim2) expression in neurons is most prominent. Adding synthetic double-stranded DNA (dsDNA) to cultured neurons induces IL-1β secretion in an AIM2-dependent manner and consequently downregulates dendritic growth but enhances axon extension. The results of Aim2 knockout and knockdown show that AIM2 acts cell-autonomously to regulate neuronal morphology. Behavioral analyses further reveal that Aim2-/- mice exhibit lower locomotor activity, increased anxious behaviors and reduced auditory fear memory. In conclusion, our study suggests that AIM2 inflammasomes regulate neuronal morphology and influence mouse behaviors.
Collapse
Affiliation(s)
- Pei-Jung Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Hsin-Yu Liu
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Yi-Ping Hsueh
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
28
|
Pohland M, Glumm R, Stoenica L, Höltje M, Kiwit J, Ahnert-Hilger G, Strauss U, Bräuer AU, Paul F, Glumm J. Studying Axonal Outgrowth and Regeneration of the Corticospinal Tract in Organotypic Slice Cultures. J Neurotrauma 2015; 32:1465-77. [DOI: 10.1089/neu.2014.3467] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Martin Pohland
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Glumm
- NeuroCure Clinical Research Center and Clinical and Experimental Research Center for Multiple Sclerosis, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Luminita Stoenica
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Höltje
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jürgen Kiwit
- Department of Neurosurgery, HELIOS Klinikum Berlin Buch, Berlin, Germany
| | - Gudrun Ahnert-Hilger
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf Strauss
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja U. Bräuer
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center and Clinical and Experimental Research Center for Multiple Sclerosis, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jana Glumm
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurosurgery, HELIOS Klinikum Berlin Buch, Berlin, Germany
| |
Collapse
|
29
|
Walsh JT, Hendrix S, Boato F, Smirnov I, Zheng J, Lukens JR, Gadani S, Hechler D, Gölz G, Rosenberger K, Kammertöns T, Vogt J, Vogelaar C, Siffrin V, Radjavi A, Fernandez-Castaneda A, Gaultier A, Gold R, Kanneganti TD, Nitsch R, Zipp F, Kipnis J. MHCII-independent CD4+ T cells protect injured CNS neurons via IL-4. J Clin Invest 2015; 125:699-714. [PMID: 25607842 PMCID: PMC4319416 DOI: 10.1172/jci76210] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 12/02/2014] [Indexed: 12/13/2022] Open
Abstract
A body of experimental evidence suggests that T cells mediate neuroprotection following CNS injury; however, the antigen specificity of these T cells and how they mediate neuroprotection are unknown. Here, we have provided evidence that T cell-mediated neuroprotection after CNS injury can occur independently of major histocompatibility class II (MHCII) signaling to T cell receptors (TCRs). Using two murine models of CNS injury, we determined that damage-associated molecular mediators that originate from injured CNS tissue induce a population of neuroprotective, IL-4-producing T cells in an antigen-independent fashion. Compared with wild-type mice, IL-4-deficient animals had decreased functional recovery following CNS injury; however, transfer of CD4+ T cells from wild-type mice, but not from IL-4-deficient mice, enhanced neuronal survival. Using a culture-based system, we determined that T cell-derived IL-4 protects and induces recovery of injured neurons by activation of neuronal IL-4 receptors, which potentiated neurotrophin signaling via the AKT and MAPK pathways. Together, these findings demonstrate that damage-associated molecules from the injured CNS induce a neuroprotective T cell response that is independent of MHCII/TCR interactions and is MyD88 dependent. Moreover, our results indicate that IL-4 mediates neuroprotection and recovery of the injured CNS and suggest that strategies to enhance IL-4-producing CD4+ T cells have potential to attenuate axonal damage in the course of CNS injury in trauma, inflammation, or neurodegeneration.
Collapse
Affiliation(s)
- James T. Walsh
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Sven Hendrix
- Department of Morphology and BIOMED Institute, Hasselt University, Diepenbeek, Belgium
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | - Francesco Boato
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Igor Smirnov
- Center for Brain Immunology and Glia
- Department of Neuroscience
| | - Jingjing Zheng
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Institute of Neurosciences, Fourth Military Medical University, Xi’an, China
| | - John R. Lukens
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sachin Gadani
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Hechler
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | - Greta Gölz
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Rosenberger
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
| | | | - Johannes Vogt
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christina Vogelaar
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Volker Siffrin
- Department of Neurology, Focus Program Translational Neuroscience and Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ali Radjavi
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Microbiology, Immunology and Infectious Diseases, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | | | - Alban Gaultier
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital/Ruhr-University Bochum, Bochum, Germany
| | | | - Robert Nitsch
- Institute for Cell Biology and Neurobiology, Center for Anatomy, Charité — Universitätsmedizin Berlin, Berlin, Germany
- Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience and Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia
- Department of Neuroscience
- Graduate Program in Neuroscience, and
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Graduate Program in Microbiology, Immunology and Infectious Diseases, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
30
|
Oncostatin M reduces lesion size and promotes functional recovery and neurite outgrowth after spinal cord injury. Mol Neurobiol 2014; 50:1142-51. [PMID: 24996996 DOI: 10.1007/s12035-014-8795-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/15/2014] [Indexed: 12/25/2022]
Abstract
The family of interleukin (IL)-6 like cytokines plays an important role in the neuroinflammatory response to injury by regulating both neural as well as immune responses. Here, we show that expression of the IL-6 family member oncostatin M (OSM) and its receptor is upregulated after spinal cord injury (SCI). To reveal the relevance of increased OSM signaling in the pathophysiology of SCI, OSM was applied locally after spinal cord hemisection in mice. OSM treatment significantly improved locomotor recovery after mild and severe SCI. Improved recovery in OSM-treated mice was associated with a reduced lesion size. OSM significantly diminished astrogliosis and immune cell infiltration. Thus, OSM limits secondary damage after CNS trauma. In vitro viability assays demonstrated that OSM protects primary neurons in culture from cell death, suggesting that the underlying mechanism involves direct neuroprotective effects of OSM. Furthermore, OSM dose-dependently promoted neurite outgrowth in cultured neurons, indicating that the cytokine plays an additional role in CNS repair. Indeed, our in vivo experiments demonstrate that OSM treatment increases plasticity of serotonergic fibers after SCI. Together, our data show that OSM is produced at the lesion site, where it protects the CNS from further damage and promotes recovery.
Collapse
|
31
|
Schizas N, Andersson B, Hilborn J, Hailer NP. Interleukin-1 receptor antagonist promotes survival of ventral horn neurons and suppresses microglial activation in mouse spinal cord slice cultures. J Neurosci Res 2014; 92:1457-65. [PMID: 24975034 DOI: 10.1002/jnr.23429] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 04/23/2014] [Accepted: 05/09/2014] [Indexed: 02/05/2023]
Abstract
Secondary damage after spinal cord injury (SCI) induces neuronal demise through neurotoxicity and inflammation, and interleukin (IL)-1β is a key inflammatory mediator. We hypothesized that IL-1β is released in spinal cord slice cultures (SCSC) and aimed at preventing the potentially neurotoxic effects of IL-1β by using interleukin-1 receptor antagonist (IL1RA). We hypothesized that IL1RA treatment enhances neuronal survival and suppresses microglial activation. SCSC were cultured up to 8 days in vitro (DIV) in the presence of IL1RA or without, either combined with trophic support using neurotrophin (NT)-3 or not. Four groups were studied: negative control, IL1RA, NT-3, and IL1RA + NT-3. IL-1β concentrations in supernatants were measured by ELISA. SCSC were immunohistochemically stained for NeuN and α-neurofilament, and microglial cells were visualized with isolectin B4 . After 8 DIV, ventral horn neurons were significantly more numerous in the IL1RA, NT-3, and IL1RA + NT-3 groups compared with negative controls. Activated microglial cells were significantly less numerous in the IL1RA, NT-3, and IL1RA + NT-3 groups compared with negative controls. Axons expanded into the collagen matrix after treatment with IL1RA, NT-3, or IL1RA + NT-3, but not in negative controls. IL-1β release from cultures peaked after 6 hr and was lowest in the IL1RA + NT-3 group. We conclude that IL-1β is released in traumatized spinal cord tissue and that IL1RA could exert its neuroprotective actions by blocking IL-1-receptors. IL1RA thereby sustains neuronal survival irrespective of the presence of additional trophic support. Microglial activation is suppressed in the presence of IL1RA, suggesting decreased inflammatory activity. IL1RA treatment approaches may have substantial impact following SCI.
Collapse
Affiliation(s)
- N Schizas
- The SpineLab, Institute of Surgical Sciences, Department of Orthopaedics, Uppsala University, SE-751 85, Uppsala, Sweden
| | | | | | | |
Collapse
|
32
|
Inflammatory profiling of Schwann cells in contact with growing axons distal to nerve injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:691041. [PMID: 24877128 PMCID: PMC4022316 DOI: 10.1155/2014/691041] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 03/28/2014] [Indexed: 11/18/2022]
Abstract
Activated Schwann cells distal to nerve injury upregulate inflammatory mediators, including cytokines. The goal of the present study was to investigate expression of proinflammatory (IL-1β, TNFα) and anti-inflammatory cytokines (IL-4, IL-10) in activated Schwann cells in relation to growing axons distal to crush injury of rat sciatic nerves. Seven days from sciatic nerve crush, transverse cryostat sections were cut 5 mm distal to lesion and incubated for double immunostaining to indicate Schwann cells (GFAP or S100b) and individual investigated cytokines or to demonstrate growing axons (GAP43). The Schwann cells of naïve sciatic nerves and those removed from sham-operated rats displayed similar weak immunoreactivity for the investigated cytokines. In contrast, increased intensity of cytokine immunofluorescence was found in Schwann cells distal to crush lesion. The cytokine-positive Schwann cells were found in close contact with growing axons detected by immunostaining for GAP43. The results of immunohistochemical analysis distal to nerve crush injury suggest that inflammatory profiling of Schwann cells including upregulation of both pro- and anti-inflammatory cytokines does not prevent growth of axons distal to nerve crush injury.
Collapse
|
33
|
Fuentes-Santamaría V, Alvarado JC, Gabaldón-Ull MC, Manuel Juiz J. Upregulation of insulin-like growth factor and interleukin 1β occurs in neurons but not in glial cells in the cochlear nucleus following cochlear ablation. J Comp Neurol 2014; 521:3478-99. [PMID: 23681983 DOI: 10.1002/cne.23362] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 12/30/2022]
Abstract
One of the main mechanisms used by neurons and glial cells to promote repair following brain injury is to upregulate activity-dependent molecules such as insulin-like growth factor 1 (IGF-1) and interleukin-1β (IL-1β). In the auditory system, IGF-1 is crucial for restoring synaptic transmission following hearing loss; however, whether IL-1β is also involved in this process is unknown. In this study, we evaluated the expression of IGF-1 and IL-1β within neurons and glial cells of the ventral cochlear nucleus in adult rats at 1, 7, 15, and 30 days following bilateral cochlear ablation. After the lesion, significant increases in both the overall mean gray levels of IGF-1 immunostaining and the mean gray levels within cells of the cochlear nucleus were observed at 1, 7, and 15 days compared with control animals. The expression and distribution of IL-1β in the ventral cochlear nucleus of ablated animals was temporally and spatially correlated with IGF-1. We also observed a lack of colocalization between IGF-1 and IL-1β with either astrocytes or microglia at any of the time points following ablation. These results suggest that the upregulation of IGF-1 and IL-1β levels within neurons-but not within glial cells-may reflect a plastic mechanism involved in repairing synaptic homeostasis of the overall cellular environment of the cochlear nucleus following bilateral cochlear ablation.
Collapse
Affiliation(s)
- Verónica Fuentes-Santamaría
- Institute for Research on Neurological Disorders (IDINE), Faculty of Medicine, University of Castilla-La Mancha, 02006, Albacete, Spain
| | | | | | | |
Collapse
|
34
|
Dooley D, Vidal P, Hendrix S. Immunopharmacological intervention for successful neural stem cell therapy: New perspectives in CNS neurogenesis and repair. Pharmacol Ther 2013; 141:21-31. [PMID: 23954656 DOI: 10.1016/j.pharmthera.2013.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/11/2022]
Abstract
The pharmacological support and stimulation of endogenous and transplanted neural stem cells (NSCs) is a major challenge in brain repair. Trauma to the central nervous system (CNS) results in a distinct inflammatory response caused by local and infiltrating immune cells. This makes NSC-supported regeneration difficult due to the presence of inhibitory immune factors which are upregulated around the lesion site. The continual and dual role of the neuroinflammatory response leaves it difficult to decipher upon a single modulatory strategy. Therefore, understanding the influence of cytokines upon regulation of NSC self-renewal, proliferation and differentiation is crucial when designing therapies for CNS repair. There is a plethora of partially conflicting data in vitro and in vivo on the role of cytokines in modulating the stem cell niche and the milieu around NSC transplants. This is mainly due to the pleiotropic role of many factors. In order for cell-based therapy to thrive, treatment must be phase-specific to the injury and also be personalized for each patient, i.e. taking age, sex, neuroimmune and endocrine status as well as other key parameters into consideration. In this review, we will summarize the most relevant information concerning interleukin (IL)-1, IL-4, IL-10, IL-15, IFN-γ, the neuropoietic cytokine family and TNF-α in order to extract promising therapeutic approaches for further research. We will focus on the consequences of neuroinflammation on endogenous brain stem cells and the transplantation environment, the effects of the above cytokines on NSCs, as well as immunopharmacological manipulation of the microenvironment for potential therapeutic use.
Collapse
Affiliation(s)
- Dearbhaile Dooley
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium
| | - Pia Vidal
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium
| | - Sven Hendrix
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium.
| |
Collapse
|
35
|
Abstract
Intestinal inflammation causes initial axonal degeneration and neuronal death but subsequent axon outgrowth from surviving neurons restores innervation density to the target smooth muscle cells. Elsewhere, the pro-inflammatory cytokines TNFα and IL-1β cause neurotoxicity, leading us to test their role in promoting enteric neuron death. In a rat coculture model, TNFα or IL-1β did not affect neuron number but did promote significant neurite outgrowth to twofold that of control by 48 h, while other cytokines (e.g., IL-4, TGFβ) were without effect. TNFα or IL-1β activated the NFκB signaling pathway, and inhibition of NFκB signaling blocked the stimulation of neurite growth. However, nuclear translocation of NFκB in smooth muscle cells but not in adjacent neurons suggested a dominant role for smooth muscle cells. TNFα or IL-1β sharply increased both mRNA and protein for GDNF, while the neurotrophic effects of TNFα or IL-1β were blocked by the RET-receptor blocker vandetanib. Conditioned medium from cytokine-treated smooth muscle cells mimicked the neurotrophic effect, inferring that TNFα and IL-1β promote neurite growth through NFκB-dependent induction of glial cell line-derived neurotrophic factor (GDNF) expression in intestinal smooth muscle cells. In vivo, TNBS-colitis caused early nuclear translocation of NFκB in smooth muscle cells. Conditioned medium from the intact smooth muscle of the inflamed colon caused a 2.5-fold increase in neurite number in cocultures, while Western blotting showed a substantial increase in GDNF protein. Pro-inflammatory cytokines promote neurite growth through upregulation of GDNF, a novel process that may facilitate re-innervation of smooth muscle cells and a return to homeostasis following initial damage.
Collapse
|
36
|
Bell K, Gramlich OW, Von Thun Und Hohenstein-Blaul N, Beck S, Funke S, Wilding C, Pfeiffer N, Grus FH. Does autoimmunity play a part in the pathogenesis of glaucoma? Prog Retin Eye Res 2013; 36:199-216. [PMID: 23541978 DOI: 10.1016/j.preteyeres.2013.02.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 02/17/2013] [Accepted: 02/18/2013] [Indexed: 12/12/2022]
Abstract
Glaucoma is a chronic neurodegenerative disease and one of the leading causes of blindness. Several risk factors have been described, e.g. an elevated intraocular pressure (IOP), oxidative stress or mitochondrial dysfunction. Additionally, alterations in serum antibody profiles of glaucoma patients, upregulation (e.g. anti-HSP60, anti-MBP) and downregulation (e.g. anti-14-3-3), have been described, but it still remains elusive if the autoantibodies seen in glaucoma are an epiphenomenon or causative. However, it is known that elicited autoimmunity causes retinal ganglion cell loss resulting in glaucomatous-like damage and according to the autoaggressive nature of some autoantibodies we found antibody deposits in human glaucomatous retinae in a pro-inflammatory environment. Furthermore, glaucomatous serum has the potential to influence neuroretinal cell regulatory processes. Importantly, we demonstrate that some autoantibodies hold neuroprotective potential for neuroretinal cells. The protective nature of autoantibodies and the molecular mechanisms underlying the very sensitive equilibrium between autoaggression and protection remain subject of future examinations and offer promising target sites for new therapeutic approaches. Additionally, the changes in antibody profiles could be used as highly sensitive and specific marker for diagnostics purposes. Early diagnosis and intervention in risk patients would offer the chance of early treatment and to slow down the progression of glaucoma and delay the resulting blindness.
Collapse
Affiliation(s)
- Katharina Bell
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutewnberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Boato F, Rosenberger K, Nelissen S, Geboes L, Peters EM, Nitsch R, Hendrix S. Absence of IL-1β positively affects neurological outcome, lesion development and axonal plasticity after spinal cord injury. J Neuroinflammation 2013; 10:6. [PMID: 23317037 PMCID: PMC3585738 DOI: 10.1186/1742-2094-10-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 12/07/2012] [Indexed: 11/10/2022] Open
Abstract
Precise crosstalk between the nervous and immune systems is important for neuroprotection and axon plasticity after injury. Recently, we demonstrated that IL-1β acts as a potent inducer of neurite outgrowth from organotypic brain slices in vitro, suggesting a potential function of IL-1β in axonal plasticity. Here, we have investigated the effects of IL-1β on axon plasticity during glial scar formation and on functional recovery in a mouse model of spinal cord compression injury (SCI). We used an IL-1β deficiency model (IL-1βKO mice) and administered recombinant IL-1β. In contrast to our hypothesis, the histological analysis revealed a significantly increased lesion width and a reduced number of corticospinal tract fibers caudal to the lesion center after local application of recombinant IL-1β. Consistently, the treatment significantly worsened the neurological outcome after SCI in mice compared with PBS controls. In contrast, the absence of IL-1β in IL-1βKO mice significantly improved recovery from SCI compared with wildtype mice. Histological analysis revealed a smaller lesion size, reduced lesion width and greatly decreased astrogliosis in the white matter, while the number of corticospinal tract fibers increased significantly 5 mm caudal to the lesion in IL-1βKO mice relative to controls. Our study for the first time characterizes the detrimental effects of IL-1β not only on lesion development (in terms of size and glia activation), but also on the plasticity of central nervous system axons after injury.
Collapse
Affiliation(s)
- Francesco Boato
- Department of Morphology & BIOMED Institute, Campus Diepenbeek, Hasselt University, Agoralaan Gebouw C, Diepenbeek, BE 3590, Belgium
| | | | | | | | | | | | | |
Collapse
|
38
|
Role of inflammation and cytokines in peripheral nerve regeneration. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 108:173-206. [PMID: 24083435 DOI: 10.1016/b978-0-12-410499-0.00007-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter provides a review of immune reactions involved in classic as well as alternative methods of peripheral nerve regeneration, and mainly with a view to understanding their beneficial effects. Axonal degeneration distal to nerve damage triggers a cascade of inflammatory events alongside injured nerve fibers known as Wallerian degeneration (WD). The early inflammatory reactions of WD comprise the complement system, arachidonic acid metabolites, and inflammatory mediators that are related to myelin fragmentation and activation of Schwann cells. Fine-tuned upregulation of the cytokine/chemokine network by Schwann cells activates resident and hematogenous macrophages to complete the clearance of axonal and myelin debris and stimulate regrowth of axonal sprouts. In addition to local effects, immune reactions of neuronal bodies and glial cells are also implicated in the survival and conditioning of neurons to regenerate severed nerves. Understanding of the cellular and molecular interactions between the immune system and peripheral nerve injury opens new possibilities for targeting inflammatory mediators to improve functional reinnervation.
Collapse
|