1
|
Li H, Yu W, Zheng X, Zhu Z. TREM1-Microglia crosstalk: Neurocognitive disorders. Brain Res Bull 2024; 220:111162. [PMID: 39645047 DOI: 10.1016/j.brainresbull.2024.111162] [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: 08/26/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Neurocognitive Disorders (NCDs) primarily affect cognitive functions, including learning, memory, perception, and problem-solving. They predominantly arise as pathological sequelae of central nervous system (CNS) disorders. Emerging evidence suggests that microglial inflammatory activation within the hippocampus underlies the pathogenesis of cognitive impairment. Triggering receptor expressed on myeloid cells 1 (TREM1), a pattern-recognition receptor on microglia, becomes upregulated in response to injury and synergistically amplifies inflammatory responses mediated by other pattern-recognition receptors, leading to uncontrolled inflammation. While TREM1 is lowly expressed in the resting state, its upregulation upon exposure to injurious inflammatory stimuli promotes microglial activation and contributes to the development of NCDs. Consequently, TREM1 may serve as a critical receptor in microglia-mediated inflammation. This article reviews the current understanding of TREM1 and its role in NCDs pathogenesis.
Collapse
Affiliation(s)
- Huashan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; Department of Anesthesiology, Zunyi Maternal And Child Health Care Hospital, Zunyi 563000, China.
| | - Wanqiu Yu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Xue Zheng
- Department of Anesthesiology, Zunyi Maternal And Child Health Care Hospital, Zunyi 563000, China
| | - Zhaoqiong Zhu
- Early Clinical Research Ward, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China.
| |
Collapse
|
2
|
Sarmin N, Roknuzzaman ASM, Sarker R, Rashid MO, Hasan A, Qusar MMAS, Kabir ER, Islam MR, Mahmud ZA. Exploring the role of interleukin-1β and interleukin-6 in the pathophysiology of obsessive-compulsive disorder. PLoS One 2024; 19:e0306125. [PMID: 38924009 PMCID: PMC11207128 DOI: 10.1371/journal.pone.0306125] [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: 01/14/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Obsessive-compulsive disorder (OCD) is a highly prevalent neuropsychiatric disorder. Recently, there has been a growing interest in investigating the association between pro-inflammatory cytokines and the pathogenesis of OCD. However, studies targeting interleukin-1β (IL-1β) and interleukin-6 (IL-6) in OCD are limited. Therefore, the present study aimed to explore the potential role of pro-inflammatory cytokines IL-1β and IL-6 in the pathophysiology and development of OCD. METHODS This study recruited 58 OCD patients and 30 age-sex-matched healthy controls (HCs). A qualified psychiatrist diagnosed OCD patients and assessed HCs based on the Diagnostic and Statistical Manual for Mental Health Disorders, 5th edition (DSM-5) criteria. We measured the severity of OCD using the Yale-Brown Obsessive Compulsive Scale (Y-BOCS). Serum IL-1β and IL-6 levels were measured using ELISA kits following the appropriate methods. RESULTS The results showed that serum IL-1β levels were significantly elevated in OCD patients compared to HCs (23.68±1.65 pg/ml vs. 15.75±1.02 pg/ml; p = 0.002). Similarly, OCD patients exhibited significantly higher serum IL-6 levels than HCs (44.97±0.73 pg/ml vs. 37.04±0.35 pg/ml; p<0.001). We observed both cytokines were positively correlated with the Y-BOCS scores in OCD patients (IL-1β: r = 0.380, p = 0.015; IL-6: r = 0.324, p = 0.026) which indicates their role in disease pathophysiology. CONCLUSION These results suggest that serum IL-1β and IL-6 levels may be associated with the pathophysiology of OCD. Also, these cytokines levels in blood samples can serve as early risk assessment tools for the development of OCD. We recommend further studies in a large and homogeneous population to support these findings.
Collapse
Affiliation(s)
- Nisat Sarmin
- Faculty of Pharmacy, Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka, Bangladesh
| | | | - Rapty Sarker
- Department of Pharmacy, University of Asia Pacific, Dhaka, Bangladesh
| | - Mamun-or- Rashid
- Faculty of Pharmacy, Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka, Bangladesh
| | - Ahasanul Hasan
- Faculty of Pharmacy, Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka, Bangladesh
| | | | | | | | - Zobaer Al Mahmud
- Faculty of Pharmacy, Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
3
|
Assis-de-Lemos G, Moura-do-Nascimento R, Amaral-do-Nascimento M, Miceli AC, Vieira TCRG. Interactions between Cytokines and the Pathogenesis of Prion Diseases: Insights and Implications. Brain Sci 2024; 14:413. [PMID: 38790392 PMCID: PMC11117815 DOI: 10.3390/brainsci14050413] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/26/2024] Open
Abstract
Transmissible Spongiform Encephalopathies (TSEs), including prion diseases such as Bovine Spongiform Encephalopathy (Mad Cow Disease) and variant Creutzfeldt-Jakob Disease, pose unique challenges to the scientific and medical communities due to their infectious nature, neurodegenerative effects, and the absence of a cure. Central to the progression of TSEs is the conversion of the normal cellular prion protein (PrPC) into its infectious scrapie form (PrPSc), leading to neurodegeneration through a complex interplay involving the immune system. This review elucidates the current understanding of the immune response in prion diseases, emphasizing the dual role of the immune system in both propagating and mitigating the disease through mechanisms such as glial activation, cytokine release, and blood-brain barrier dynamics. We highlight the differential cytokine profiles associated with various prion strains and stages of disease, pointing towards the potential for cytokines as biomarkers and therapeutic targets. Immunomodulatory strategies are discussed as promising avenues for mitigating neuroinflammation and delaying disease progression. This comprehensive examination of the immune response in TSEs not only advances our understanding of these enigmatic diseases but also sheds light on broader neuroinflammatory processes, offering hope for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Tuane C. R. G. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis and National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.A.-d.-L.); (R.M.-d.-N.); (M.A.-d.-N.); (A.C.M.)
| |
Collapse
|
4
|
Lindblad C, Rostami E, Helmy A. Interleukin-1 Receptor Antagonist as Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1508-1528. [PMID: 37610701 PMCID: PMC10684479 DOI: 10.1007/s13311-023-01421-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury is a common type of acquired brain injury of varying severity carrying potentially deleterious consequences for the afflicted individuals, families, and society. Following the initial, traumatically induced insult, cellular injury processes ensue. These are believed to be amenable to treatment. Among such injuries, neuroinflammation has gained interest and has become a specific focus for both experimental and clinical researchers. Neuroinflammation is elicited almost immediately following trauma, and extend for a long time, possibly for years, after the primary injury. In the acute phase, the inflammatory response is characterized by innate mechanisms such as the activation of microglia which among else mediates cytokine production. Among the earliest cytokines to emerge are the interleukin- (IL-) 1 family members, comprising, for example, the agonist IL-1β and its competitive antagonist, IL-1 receptor antagonist (IL-1ra). Because of its early emergence following trauma and its increased concentrations also after human TBI, IL-1 has been hypothesized to be a tractable treatment target following TBI. Ample experimental data supports this, and demonstrates restored neurological behavior, diminished lesion zones, and an attenuated inflammatory response following IL-1 modulation either through IL-1 knock-out experiments, IL-1β inhibition, or IL-1ra treatment. Of these, IL-1ra treatment is likely the most physiological. In addition, recombinant human IL-1ra (anakinra) is already approved for utilization across a few rheumatologic disorders. As of today, one randomized clinical controlled trial has utilized IL-1ra inhibition as an intervention and demonstrated its safety. Further clinical trials powered for patient outcome are needed in order to demonstrate efficacy. In this review, we summarize IL-1 biology in relation to acute neuroinflammatory processes following TBI with a particular focus on current evidence for IL-1ra treatment both in the experimental and clinical context.
Collapse
Affiliation(s)
- Caroline Lindblad
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Elham Rostami
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Fan H, Sheng S, Li D, Li J, Wang G, Zhang F. Heat-killed Lactobacillus murinus confers neuroprotection against dopamine neuronal loss by targeting NLRP3 inflammasome. Bioeng Transl Med 2023; 8:e10455. [PMID: 36925673 PMCID: PMC10013757 DOI: 10.1002/btm2.10455] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/12/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
The intestinal flora has become very active in studies related to Parkinson's disease (PD) in recent years. The microbe-gut-brain axis is closely related to the maintenance of brain homeostasis as well as PD pathogenesis. Alterations in gut bacteria can contribute to neuroinflammation and dopamine (DA) neurodegeneration. Lactobacillus murinus, a gram-positive bacterium, is a commensal gut bacteria present in the mammalian gut and considered as a potential probiotic due to its beneficial effects, including anti-inflammatory and antibacterial actions. In this study, the effects of live L. murinus and heat-killed L. murinus on DA neuronal damage in rats and the underlying mechanisms were investigated. Data showed that heat-killed L. murinus ameliorated 6-hydroxydopamine-induced motor dysfunctions and loss of substantia nigra DA neurons, while no protection was shown in live L. murinus treatment. At the same time, heat-killed L. murinus reduced the activation of NLRP3 inflammasome in microglia and the secretion of pro-inflammatory factors, thus inhibiting the development of neuroinflammation. Furthermore, heat-killed L. murinus failed to display its original neuroprotective properties in NLRP3 inflammasome knockout mice. Together, heat-killed L. murinus conferred neuroprotection against DA neuronal loss via the inhibition of microglial NLRP3 inflammasome activation. These findings provide a promising potential for future applications of L. murinus, and also beneficial strategy for PD treatment.
Collapse
Affiliation(s)
- Hong‐Xia Fan
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal CenterZunyi Medical UniversityZunyiGuizhouChina
| | - Shuo Sheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal CenterZunyi Medical UniversityZunyiGuizhouChina
| | - Dai‐Di Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal CenterZunyi Medical UniversityZunyiGuizhouChina
| | - Jing‐Jie Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal CenterZunyi Medical UniversityZunyiGuizhouChina
| | - Guo‐Qing Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal CenterZunyi Medical UniversityZunyiGuizhouChina
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal CenterZunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
6
|
Crosstalk between PI3K/AKT/KLF4 signaling and microglia M1/M2 polarization as a novel mechanistic approach towards flibanserin repositioning in parkinson's disease. Int Immunopharmacol 2022; 112:109191. [PMID: 36055034 DOI: 10.1016/j.intimp.2022.109191] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 02/07/2023]
Abstract
Balancing microglia M1/M2 polarization has been shown as a prospective therapeutic strategy for Parkinson's disease (PD). Various vital signaling pathways are likely to govern the microglial phenotype. The implication of 5HT1A receptors in neurodegenerative disorders has raised interest in exploring the repositioning of flibanserin (Flib), a 5HT1A agonist, as an effective neuroprotective agent for PD. Therefore, this study was designed to assess the ability of Flib to modulate microglia phenotype switching from M1 to M2 via PI3K/AKT downstream targets in a rotenone model of PD. Rats received rotenone (1.5 mg/kg) every other day and were concurrently treated with Flib (40 mg/kg/day) with or without wortmannin (15 μg/kg/day), a PI3K inhibitor, for 21 days. Flib improved the motor perturbations induced by rotenone, as confirmed by the reversion of histopathological damage and tyrosine hydroxylase immunohistochemical alterations in both the striata and substantia nigra. The molecular signaling of Flib was elaborated by inducing striatal AKT phosphorylation and the expression of its substantial target, KLF4. Flib induced STAT6 phosphorylation to promote M2 polarization as demonstrated by the increased CD163++ microglial count with striatal arginase activity. In parallel, it markedly inhibited M1 activation as evidenced by the reduction in CD86++ microglia count with striatal proinflammatory mediators, IL-1β and iNOS. The pre-administration of wortmannin mostly negated Flib's neuroprotective effects. In conclusion, Flib AKT/ KLF4-dependently amended M1/M2 microglial imbalance to exert a promising neuroprotective effect, highlighting its potential as a revolutionary candidate for conquering PD.
Collapse
|
7
|
Kalra S, Malik R, Singh G, Bhatia S, Al-Harrasi A, Mohan S, Albratty M, Albarrati A, Tambuwala MM. Pathogenesis and management of traumatic brain injury (TBI): role of neuroinflammation and anti-inflammatory drugs. Inflammopharmacology 2022; 30:1153-1166. [PMID: 35802283 PMCID: PMC9293826 DOI: 10.1007/s10787-022-01017-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 02/08/2023]
Abstract
Traumatic brain injury (TBI) is an important global health concern that represents a leading cause of death and disability. It occurs due to direct impact or hit on the head caused by factors such as motor vehicles, crushes, and assaults. During the past decade, an abundance of new evidence highlighted the importance of inflammation in the secondary damage response that contributes to neurodegenerative and neurological deficits after TBI. It results in disruption of the blood-brain barrier (BBB) and initiates the release of macrophages, neutrophils, and lymphocytes at the injury site. A growing number of researchers have discovered various signalling pathways associated with the initiation and progression of inflammation. Targeting different signalling pathways (NF-κB, JAK/STAT, MAPKs, PI3K/Akt/mTOR, GSK-3, Nrf2, RhoGTPase, TGF-β1, and NLRP3) helps in the development of novel anti-inflammatory drugs in the management of TBI. Several synthetic and herbal drugs with both anti-inflammatory and neuroprotective potential showed effective results. This review summarizes different signalling pathways, associated pathologies, inflammatory mediators, pharmacological potential, current status, and challenges with anti-inflammatory drugs.
Collapse
Affiliation(s)
- Sunishtha Kalra
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Rohit Malik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India.
| | - Saurabh Bhatia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India. .,Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman.
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Syam Mohan
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.,Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ali Albarrati
- Rehabilitation Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Northern Ireland, UK.
| |
Collapse
|
8
|
Oyeleke MB, Owoyele BV. Saponins and flavonoids from Bacopa floribunda plant extract exhibit antioxidant and anti-inflammatory effects on amyloid beta 1-42-induced Alzheimer's disease in BALB/c mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114997. [PMID: 35033624 DOI: 10.1016/j.jep.2022.114997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/30/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bacopa floribunda (BF), a locally available plant has been employed traditionally as memory enhancer in Southwestern, Nigeria. It has been utilized in traditional and Ayurvedic medicine as brain tonic for enhancing memory, anti-aging and forestalling series of psychological disorders. However, there is a dearth of scientific information on the mechanism(s) of action of important phytochemicals from BF extract on dementia. AIM OF THE STUDY Alzheimer's disease, the commonest form of dementia has been postulated to triple by 2050 as a result of increase in life expectancy. This study therefore assessed and compared the possible mechanism(s) of action of flavonoids and saponins from BF on Amyloid beta (Aβ1-42)-induced dementia in male BALB/c mice. MATERIALS AND METHODS Eighty (80) healthy BALB/c mice divided into 10 groups (n = 8) were given a single bilateral ICV injection of Aβ1-42 or normal saline. Graded doses of Saponins and flavonoids (50, 100 and 200 mg/kg) were used as treatment for 21 days. Hippocampal homogenates were assayed for the levels of antioxidants, oxidative stress and neuroinflammatory markers. In vitro antioxidant activity of flavonoids and saponins were equally assessed using standard procedures. The extent of microglial activation was quantified through immunohistochemistry procedure. RESULTS Aβ1-42 successfully caused a spike in hippocampal levels of MDA, IL1β, TNF-α including MPO levels and invariably decreased antioxidant activities. Likewise an increase in reactive microglia (microgliosis) was observed. However, crude saponins and flavonoids from BF were able to suppress microgliosis, oxidative stress and neuroinflammation induced by Aβ1- 42 and were observed to be more effective at higher doses of saponins (100 mg/kg and 200 mg/kg) and flavonoid (100 mg/kg). CONCLUSIONS Phytochemicals from BF efficiently exhibited dose dependent alleviation of some symptoms associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Mosunmola Busayo Oyeleke
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine and Health Sciences, Afe Babalola University, P.M.B, 5454, Ado-Ekiti, Nigeria; Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, P.M.B, 1515, Ilorin, Nigeria.
| | - Bamidele Victor Owoyele
- Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, P.M.B, 1515, Ilorin, Nigeria.
| |
Collapse
|
9
|
Pilat D, Paumier JM, García-González L, Louis L, Stephan D, Manrique C, Khrestchatisky M, Di Pasquale E, Baranger K, Rivera S. MT5-MMP promotes neuroinflammation, neuronal excitability and Aβ production in primary neuron/astrocyte cultures from the 5xFAD mouse model of Alzheimer’s disease. J Neuroinflammation 2022; 19:65. [PMID: 35277173 PMCID: PMC8915472 DOI: 10.1186/s12974-022-02407-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/30/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Membrane-type matrix metalloproteinase 5 (MT5-MMP) deficiency in the 5xFAD mouse model of Alzheimer's disease (AD) reduces brain neuroinflammation and amyloidosis, and prevents deficits in synaptic activity and cognition in prodromal stages of the disease. In addition, MT5-MMP deficiency prevents interleukin-1 beta (IL-1β)-mediated inflammation in the peripheral nervous system. In this context, we hypothesized that the MT5-MMP/IL-1β tandem could regulate nascent AD pathogenic events in developing neural cells shortly after the onset of transgene activation.
Methods
To test this hypothesis, we used 11–14 day in vitro primary cortical cultures from wild type, MT5-MMP−/−, 5xFAD and 5xFAD/MT5-MMP−/− mice, and evaluated the impact of MT5-MMP deficiency and IL-1β treatment for 24 h, by performing whole cell patch-clamp recordings, RT-qPCR, western blot, gel zymography, ELISA, immunocytochemistry and adeno-associated virus (AAV)-mediated transduction.
Results
5xFAD cells showed higher levels of MT5-MMP than wild type, concomitant with higher basal levels of inflammatory mediators. Moreover, MT5-MMP-deficient cultures had strong decrease of the inflammatory response to IL-1β, as well as decreased stability of recombinant IL-1β. The levels of amyloid beta peptide (Aβ) were similar in 5xFAD and wild-type cultures, and IL-1β treatment did not affect Aβ levels. Instead, the absence of MT5-MMP significantly reduced Aβ by more than 40% while sparing APP metabolism, suggesting altogether no functional crosstalk between IL-1β and APP/Aβ, as well as independent control of their levels by MT5-MMP. The lack of MT5-MMP strongly downregulated the AAV-induced neuronal accumulation of the C-terminal APP fragment, C99, and subsequently that of Aβ. Finally, MT5-MMP deficiency prevented basal hyperexcitability observed in 5xFAD neurons, but not hyperexcitability induced by IL-1β treatment.
Conclusions
Neuroinflammation and hyperexcitability precede Aβ accumulation in developing neural cells with nascent expression of AD transgenes. MT5-MMP deletion is able to tune down basal neuronal inflammation and hyperexcitability, as well as APP/Aβ metabolism. In addition, MT5-MMP deficiency prevents IL-1β-mediated effects in brain cells, except hyperexcitability. Overall, this work reinforces the idea that MT5-MMP is at the crossroads of pathogenic AD pathways that are already incipiently activated in developing neural cells, and that targeting MT5-MMP opens interesting therapeutic prospects.
Collapse
|
10
|
Manjally AV, Tay TL. Attack of the Clones: Microglia in Health and Disease. Front Cell Neurosci 2022; 16:831747. [PMID: 35173585 PMCID: PMC8841846 DOI: 10.3389/fncel.2022.831747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Affiliation(s)
- Amritha Vinayak Manjally
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Centre, University of Freiburg, Freiburg, Germany
- Department of Biology, Boston University, Boston, MA, United States
| | - Tuan Leng Tay
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Centre, University of Freiburg, Freiburg, Germany
- Department of Biology, Boston University, Boston, MA, United States
- Freiburg Institute of Advanced Studies, University of Freiburg, Freiburg, Germany
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
- *Correspondence: Tuan Leng Tay
| |
Collapse
|
11
|
Allison RL, Welby E, Khayrullina G, Burnett BG, Ebert AD. Viral mediated knockdown of GATA6 in SMA iPSC-derived astrocytes prevents motor neuron loss and microglial activation. Glia 2022; 70:989-1004. [PMID: 35088910 PMCID: PMC9303278 DOI: 10.1002/glia.24153] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
Abstract
Spinal muscular atrophy (SMA), a pediatric genetic disorder, is characterized by the profound loss of spinal cord motor neurons and subsequent muscle atrophy and death. Although the mechanisms underlying motor neuron loss are not entirely clear, data from our work and others support the idea that glial cells contribute to disease pathology. GATA6, a transcription factor that we have previously shown to be upregulated in SMA astrocytes, is negatively regulated by SMN (survival motor neuron) and can increase the expression of inflammatory regulator NFκB. In this study, we identified upregulated GATA6 as a contributor to increased activation, pro-inflammatory ligand production, and neurotoxicity in spinal-cord patterned astrocytes differentiated from SMA patient induced pluripotent stem cells. Reducing GATA6 expression in SMA astrocytes via lentiviral infection ameliorated these effects to healthy control levels. Additionally, we found that SMA astrocytes contribute to SMA microglial phagocytosis, which was again decreased by lentiviral-mediated knockdown of GATA6. Together these data identify a role of GATA6 in SMA astrocyte pathology and further highlight glia as important targets of therapeutic intervention in SMA.
Collapse
Affiliation(s)
- Reilly L Allison
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Emily Welby
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Guzal Khayrullina
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland, USA
| | - Barrington G Burnett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
12
|
Sjöström EO, Culot M, Leickt L, Åstrand M, Nordling E, Gosselet F, Kaiser C. Transport study of interleukin-1 inhibitors using a human in vitro model of the blood-brain barrier. Brain Behav Immun Health 2021; 16:100307. [PMID: 34589799 PMCID: PMC8474601 DOI: 10.1016/j.bbih.2021.100307] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/24/2021] [Indexed: 01/15/2023] Open
Abstract
The proinflammatory cytokine Interleukin-1 (IL-1), with its two isoforms α and β, has important roles in multiple pathogenic processes in the central nervous system. The present study aimed to evaluate and compare the blood-to-brain distribution of anakinra (IL-1 receptor antagonist), bermekimab (IL-1α antagonist) and canakinumab (IL-1β antagonist). A human in vitro model of the blood-brain barrier derived from human umbilical cord blood stem cells was used, where isolated CD34+ cells co-cultured with bovine pericytes were matured into polarized brain-like endothelial cells. Transport rates of the three test items were evaluated after 180 min incubation at concentrations 50, 250 and 1250 nM in a transwell system. We report herein that anakinra passes the human brain-like endothelial monolayer at a 4-7-fold higher rate than the monoclonal antibodies tested. Both antibodies had similar transport rates at all concentrations. No dose-dependent effects in transport rates were observed, nor any saturation effects at supraphysiological concentrations. The larger propensity of anakinra to pass this model of the human blood-brain barrier supports existing data and confirms that anakinra can reach the brain compartment at clinically relevant concentrations. As anakinra inhibits the actions of both IL-1α and IL-1β, it blocks all effects of IL-1 downstream signaling. The results herein further add to the growing body of evidence of the potential utility of anakinra to treat neuroinflammatory disorders. Anakinra has a larger propensity to pass the in vitro BBB than monoclonal antibodies targeting the IL-1 system. Implications for targeting inflammation in cerebral ischemia and neurological sequelae of autoinflammatory diseases. Novel and comparative study of biologics in a human in vitro BBB model shows relevance and validity.
Collapse
Affiliation(s)
| | - Maxime Culot
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Lisa Leickt
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Mikael Åstrand
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Erik Nordling
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Fabien Gosselet
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Christina Kaiser
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| |
Collapse
|
13
|
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
|
14
|
Siebold L, Krueger AC, Abdala JA, Figueroa JD, Bartnik-Olson B, Holshouser B, Wilson CG, Ashwal S. Cosyntropin Attenuates Neuroinflammation in a Mouse Model of Traumatic Brain Injury. Front Mol Neurosci 2020; 13:109. [PMID: 32670020 PMCID: PMC7332854 DOI: 10.3389/fnmol.2020.00109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Abstract
Aim: Traumatic brain injury (TBI) is a leading cause of mortality/morbidity and is associated with chronic neuroinflammation. Melanocortin receptor agonists including adrenocorticotropic hormone (ACTH) ameliorate inflammation and provide a novel therapeutic approach. We examined the effect of long-acting cosyntropin (CoSyn), a synthetic ACTH analog, on the early inflammatory response and functional outcome following experimental TBI. Methods: The controlled cortical impact model was used to induce TBI in mice. Mice were assigned to injury and treatment protocols resulting in four experimental groups including sham + saline, sham + CoSyn, TBI + saline, and TBI + CoSyn. Treatment was administered subcutaneously 3 h post-injury and daily injections were given for up to 7 days post-injury. The early inflammatory response was evaluated at 3 days post-injury through the evaluation of cytokine expression (IL1β and TNFα) and immune cell response. Quantification of immune cell response included cell counts of microglia/macrophages (Iba1+ cells) and neutrophils (MPO+ cells) in the cortex and hippocampus. Behavioral testing (n = 10–14 animals/group) included open field (OF) and novel object recognition (NOR) during the first week following injury and Morris water maze (MWM) at 10–15 days post-injury. Results: Immune cell quantification showed decreased accumulation of Iba1+ cells in the perilesional cortex and CA1 region of the hippocampus for CoSyn-treated TBI animals compared to saline-treated. Reduced numbers of MPO+ cells were also found in the perilesional cortex and hippocampus in CoSyn treated TBI mice compared to their saline-treated counterparts. Furthermore, CoSyn treatment reduced IL1β expression in the cortex of TBI mice. Behavioral testing showed a treatment effect of CoSyn for NOR with CoSyn increasing the discrimination ratio in both TBI and Sham groups, indicating increased memory performance. CoSyn also decreased latency to find platform during the early training period of the MWM when comparing CoSyn to saline-treated TBI mice suggesting moderate improvements in spatial memory following CoSyn treatment. Conclusion: Reduced microglia/macrophage accumulation and neutrophil infiltration in conjunction with moderate improvements in spatial learning in our CoSyn treated TBI mice suggests a beneficial anti-inflammatory effect of CoSyn following TBI.
Collapse
Affiliation(s)
- Lorraine Siebold
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Amy C Krueger
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jonathan A Abdala
- The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Johnny D Figueroa
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brenda Bartnik-Olson
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Barbara Holshouser
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Christopher G Wilson
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States.,Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA, United States
| |
Collapse
|
15
|
Ozen I, Ruscher K, Nilsson R, Flygt J, Clausen F, Marklund N. Interleukin-1 Beta Neutralization Attenuates Traumatic Brain Injury-Induced Microglia Activation and Neuronal Changes in the Globus Pallidus. Int J Mol Sci 2020; 21:ijms21020387. [PMID: 31936248 PMCID: PMC7014296 DOI: 10.3390/ijms21020387] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/29/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) increases the risk of delayed neurodegenerative processes, including Parkinson’s disease (PD). Interleukin-1beta (IL-1β), a key pro-inflammatory cytokine, may promote secondary injury development after TBI. Conversely, neutralizing IL-1β was found to improve functional recovery following experimental TBI. However, the mechanisms underlying the behavioral improvements observed by IL-1β neutralization are still poorly understood. The present study investigated the role of IL-1β on the microglia response and neuronal changes in the globus pallidus in response to diffuse TBI. Mice were subjected to sham injury or the central fluid percussion injury (cFPI) (a model of traumatic axonal injury), and were randomly administered an IL-1β neutralizing or a control antibody at 30 min post-injury. The animals were analyzed at 2, 7, or 14 days post-injury. When compared to controls, mice subjected to cFPI TBI had increased microglia activation and dopaminergic innervation in the globus pallidus, and a decreased number of parvalbumin (PV) positive interneurons in the globus pallidus. Neutralization of IL-1β attenuated the microglia activation, prevented the loss of PV+ interneurons and normalized dopaminergic fiber density in the globus pallidus of brain-injured animals. These findings argue for an important role for neuro-inflammation in the PD-like pathology observed in TBI.
Collapse
Affiliation(s)
- Ilknur Ozen
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden; (I.O.); (K.R.); (R.N.)
| | - Karsten Ruscher
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden; (I.O.); (K.R.); (R.N.)
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden
| | - Robert Nilsson
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden; (I.O.); (K.R.); (R.N.)
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden
| | - Johanna Flygt
- Department of Neuroscience, Section of Neurosurgery, Uppsala University, 75185 Uppsala, Sweden; (J.F.); (F.C.)
| | - Fredrik Clausen
- Department of Neuroscience, Section of Neurosurgery, Uppsala University, 75185 Uppsala, Sweden; (J.F.); (F.C.)
| | - Niklas Marklund
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden; (I.O.); (K.R.); (R.N.)
- Department of Neuroscience, Section of Neurosurgery, Uppsala University, 75185 Uppsala, Sweden; (J.F.); (F.C.)
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, 22185 Lund, Sweden
- Correspondence:
| |
Collapse
|
16
|
Głombik K, Trojan E, Kurek A, Budziszewska B, Basta-Kaim A. Inflammatory Consequences of Maternal Diabetes on the Offspring Brain: a Hippocampal Organotypic Culture Study. Neurotox Res 2019; 36:357-375. [PMID: 31197747 PMCID: PMC6616224 DOI: 10.1007/s12640-019-00070-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/10/2019] [Accepted: 05/28/2019] [Indexed: 01/09/2023]
Abstract
Gestational diabetes is a disorder associated with abnormal chronic inflammation that poses a risk to the developing fetus. We investigated the effects of experimentally induced diabetes (streptozotocin model) in Wistar female rats on the inflammatory status of the hippocampi of their offspring. Additionally, the impact of antidiabetic drugs (metformin and glyburide) on inflammatory processes was evaluated. Organotypic hippocampal cultures (OHCs) were prepared from the brains of the 7-day-old rat offspring of control and diabetic mother rats. On the 7th day in vitro, the cultures were pretreated with metformin (3 μM) or glyburide (1 μM) and then stimulated for 24 h with lipopolysaccharide (LPS, 1 μg/ml). The OHCs obtained from the offspring of diabetic mothers were characterized by the increased mortality of cells and an enhanced susceptibility to damage caused by LPS. Although we showed that LPS stimulated the secretion of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) in the control and diabetic cultures, the levels of IL-1β and IL-6 in the OHC medium obtained from the offspring of diabetic mothers were more pronounced. In the diabetic cultures, enhanced levels of TLR-4 and the overactivation of the NLRP3 inflammasome were demonstrated. Metformin and glyburide pretreatment normalized the LPS-induced IL-1β secretion in the control and diabetic cultures. Furthermore, glyburide diminished both: LPS-induced IL-6 and TNF-α secretion in the control and diabetic cultures and increased NF-κB p65 subunit phosphorylation. Glyburide also diminished the levels of the NLRP3 subunit and caspase-1, but only in the diabetic cultures. The results showed that maternal diabetes affected inflammatory processes in the offspring brain and increased hippocampal sensitivity to the LPS-induced inflammatory response. The use of antidiabetic agents, especially glyburide, had a beneficial impact on the changes caused by maternal diabetes.
Collapse
Affiliation(s)
- Katarzyna Głombik
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland.
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Anna Kurek
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Bogusława Budziszewska
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| |
Collapse
|
17
|
Younger D, Murugan M, Rama Rao KV, Wu LJ, Chandra N. Microglia Receptors in Animal Models of Traumatic Brain Injury. Mol Neurobiol 2018; 56:5202-5228. [PMID: 30554385 DOI: 10.1007/s12035-018-1428-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
18
|
Herman FJ, Pasinetti GM. Principles of inflammasome priming and inhibition: Implications for psychiatric disorders. Brain Behav Immun 2018; 73:66-84. [PMID: 29902514 PMCID: PMC6526722 DOI: 10.1016/j.bbi.2018.06.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/28/2018] [Accepted: 06/09/2018] [Indexed: 12/27/2022] Open
Abstract
The production of inflammatory proteins by the innate immune system is a tightly orchestrated procedure that allows the body to efficiently respond to exogenous and endogenous threats. Recently, accumulating evidence has indicated that disturbances in the inflammatory response system not only provoke autoimmune disorders, but also can have deleterious effects on neuronal function and mental health. As inflammation in the brain is primarily mediated by microglia, there has been an expanding focus on the mechanisms through which these cells initiate and propagate neuroinflammation. Microglia can enter persistently active states upon their initial recognition of an environmental stressor and are thereafter prone to elicit amplified and persistent inflammatory responses following subsequent exposures to stressors. A recent focus on why primed microglia cells are susceptible to environmental insults has been the NLRP3 inflammasome. Its function within the innate immune system is regulated in such a manner that supports a role for the complex in gating neuroinflammatory responses. The activation of NLRP3 inflammasome in microglia results in the cleavage of zymogen inflammatory interleukins into functional forms that elicit a number of consequential effects in the local neuronal environment. There is evidence to support the principle that within primed neuroimmune systems a lowered threshold for NLRP3 activation can cause persistent neuroinflammation or the amplified production of inflammatory cytokines, such as IL-1β and IL-18. Over the course of an individual's lifetime, persistent neuroinflammation can subsequently lead to the pathophysiological signatures that define psychological disorders. Therefore, targeting the NLRP3 inflammasome complex may represent an innovative and consequential approach to limit neuroinflammatory states in psychiatric disorders, such as major depressive disorder.
Collapse
Affiliation(s)
- Francis J. Herman
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029, USA,Department of Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029, USA; Department of Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA.
| |
Collapse
|
19
|
Chung JY, Krapp N, Wu L, Lule S, McAllister LM, Edmiston WJ, Martin S, Levy E, Songtachalert T, Sherwood JS, Buckley EM, Sanders B, Izzy S, Hickman S, Guo S, Lok J, El Khoury J, Lo EH, Kaplan D, Whalen MJ. Interleukin-1 Receptor 1 Deletion in Focal and Diffuse Experimental Traumatic Brain Injury in Mice. J Neurotrauma 2018; 36:370-379. [PMID: 29768967 DOI: 10.1089/neu.2018.5659] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Important differences in the biology of focal and diffuse traumatic brain injury (TBI) subtypes may result in unique pathophysiological responses to shared molecular mechanisms. Interleukin-1 (IL-1) signaling has been tested as a potential therapeutic target in preclinical models of cerebral contusion and diffuse TBI, and in a phase II clinical trial, but no published studies have examined IL-1 signaling in an impact/acceleration closed head injury (CHI) model. We hypothesized that genetic deletion of IL-1 receptor-1 (IL-1R1 KO) would be beneficial in focal (contusion) and CHI in mice. Wild type and IL-1R1 KO mice were subjected to controlled cortical impact (CCI), or to CHI. CCI produced brain leukocyte infiltration, HMGB1 translocation and release, edema, cell death, and cognitive deficits. CHI induced peak rotational acceleration of 9.7 × 105 ± 8.1 × 104 rad/s2, delayed time to righting reflex, and robust Morris water maze deficits without deficits in tests of anxiety, locomotion, sensorimotor function, or depression. CHI produced no discernable acute plasmalemma damage or cell death, blood-brain barrier permeability to IgG, or brain edema and only a modest increase in brain leukocyte infiltration at 72 h. In both models, mature (17 kDa) interleukin-1 beta (IL-1β) was induced by 24 h in CD31+ endothelial cells isolated from injured brain but was not induced in CD11b+ cells in either model. High mobility group box protein-1 was released from injured brain cells in CCI but not CHI. Surprisingly, cognitive outcome in mice with global deletion of IL-1R1 was improved in CHI, but worse after CCI without affecting lesion size, edema, or infiltration of CD11b+/CD45+ leukocytes in CCI. IL-1R1 may induce unique biological responses, beneficial or detrimental to cognitive outcome, after TBI depending on the pathoanatomical subtype. Brain endothelium is a hitherto unrecognized source of mature IL-1β in both models.
Collapse
Affiliation(s)
- Joon Yong Chung
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nicolas Krapp
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,3 Medizinische Fakultät Mannheim, Heidelberg University, Mannheim, Germany
| | - Limin Wu
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sevda Lule
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lauren M McAllister
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - William J Edmiston
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Samantha Martin
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emily Levy
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tanya Songtachalert
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John S Sherwood
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Erin M Buckley
- 4 Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.,5 Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Bharat Sanders
- 4 Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Saef Izzy
- 6 Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Suzanne Hickman
- 7 Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuzhen Guo
- 8 Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Josephine Lok
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joseph El Khoury
- 7 Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eng H Lo
- 8 Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - David Kaplan
- 9 Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael J Whalen
- 1 Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,2 Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Swaroop S, Mahadevan A, Shankar SK, Adlakha YK, Basu A. HSP60 critically regulates endogenous IL-1β production in activated microglia by stimulating NLRP3 inflammasome pathway. J Neuroinflammation 2018; 15:177. [PMID: 29885667 PMCID: PMC5994257 DOI: 10.1186/s12974-018-1214-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/23/2018] [Indexed: 11/10/2022] Open
Abstract
Background Interleukin-1β (IL-1β) is one of the most important cytokine secreted by activated microglia as it orchestrates the vicious cycle of inflammation by inducing the expression of various other pro-inflammatory cytokines along with its own production. Microglia-mediated IL-1β production is a tightly regulated mechanism which involves the activation of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome pathway. Our previous study suggests the critical role of heat shock protein 60 (HSP60) in IL-1β-induced inflammation in microglia through TLR4-p38 MAPK axis. However, whether HSP60 regulates endogenous IL-1β production is not known. Therefore, to probe the underlying mechanism, we elucidate the role of HSP60 in endogenous IL-1β production. Methods We used in vitro (N9 murine microglial cells) and in vivo (BALB/c mouse) models for our study. HSP60 overexpression and knockdown experiment was done to elucidate the role of HSP60 in endogenous IL-1β production by microglia. Western blotting and quantitative real-time PCR was performed using N9 cells and BALB/c mice brain, to analyze various proteins and transcript levels. Reactive oxygen species levels and mitochondrial membrane depolarization in N9 cells were analyzed by flow cytometry. We also performed caspase-1 activity assay and enzyme-linked immunosorbent assay to assess caspase-1 activity and IL-1β production, respectively. Results HSP60 induces the phosphorylation and nuclear localization of NF-κB both in vitro and in vivo. It also induces perturbation in mitochondrial membrane potential and enhances reactive oxygen species (ROS) generation in microglia. HSP60 further activates NLRP3 inflammasome by elevating NLRP3 expression both at RNA and protein levels. Furthermore, HSP60 enhances caspase-1 activity and increases IL-1β secretion by microglia. Knockdown of HSP60 reduces the IL-1β-induced production of IL-1β both in vitro and in vivo. Also, we have shown for the first time that knockdown of HSP60 leads to decreased IL-1β production during Japanese encephalitis virus (JEV) infection, which eventually leads to decreased inflammation and increased survival of JEV-infected mice. Conclusion HSP60 mediates microglial IL-1β production by regulating NLRP3 inflammasome pathway and reduction of HSP60 leads to reduction of inflammation in JEV infection. Electronic supplementary material The online version of this article (10.1186/s12974-018-1214-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shalini Swaroop
- National Brain Research Centre, Manesar, Haryana, 122052, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Yogita K Adlakha
- National Brain Research Centre, Manesar, Haryana, 122052, India.
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, 122052, India.
| |
Collapse
|
21
|
Microsomal Prostaglandin E Synthase-1 Facilitates an Intercellular Interaction between CD4⁺ T Cells through IL-1β Autocrine Function in Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2017; 18:ijms18122758. [PMID: 29257087 PMCID: PMC5751357 DOI: 10.3390/ijms18122758] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/09/2017] [Accepted: 12/13/2017] [Indexed: 11/21/2022] Open
Abstract
Microsomal prostaglandin synthetase-1 (mPGES-1) is an inducible terminal enzyme that produces prostaglandin E2 (PGE2). In our previous study, we investigated the role of mPGES-1 in the inflammation and demyelination observed in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, using mPGES-1-deficient (mPGES-1−/−) and wild-type (wt) mice. We found that mPGES-1 facilitated inflammation, demyelination, and paralysis and was induced in vascular endothelial cells and macrophages and microglia around inflammatory foci. Here, we investigated the role of interleukin-1β (IL-1β) in the intercellular mechanism stimulated by mPGES-1 in EAE spinal cords in the presence of inflammation. We found that the area invaded by CD4-positive (CD4+) T cells was extensive, and that PGE2 receptors EP1–4 were more induced in activated CD4+ T cells of wt mice than in those of mPGES-1−/− mice. Moreover, IL-1β and IL-1 receptor 1 (IL-1r1) were produced by 65% and 48% of CD4+ T cells in wt mice and by 44% and 27% of CD4+ T cells in mPGES-1−/− mice. Furthermore, interleukin-17 (IL-17) was released from the activated CD4+ T cells. Therefore, mPGES-1 stimulates an intercellular interaction between CD4+ T cells by upregulating the autocrine function of IL-1β in activated CD4+ T cells, which release IL-17 to facilitate axonal and myelin damage in EAE mice.
Collapse
|
22
|
Anti-inflammatory (M2) macrophage media reduce transmission of oligomeric amyloid beta in differentiated SH-SY5Y cells. Neurobiol Aging 2017; 60:173-182. [DOI: 10.1016/j.neurobiolaging.2017.08.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/16/2017] [Accepted: 08/19/2017] [Indexed: 01/28/2023]
|
23
|
Hutson LW, Lebonville CL, Jones ME, Fuchs RA, Lysle DT. Interleukin-1 signaling in the basolateral amygdala is necessary for heroin-conditioned immunosuppression. Brain Behav Immun 2017; 62:171-179. [PMID: 28131792 PMCID: PMC5828772 DOI: 10.1016/j.bbi.2017.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/14/2017] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
Heroin administration suppresses the production of inducible nitric oxide (NO), as indicated by changes in splenic inducible nitric oxide synthase (iNOS) and plasma nitrate/nitrite. Since NO is a measure of host defense against infection and disease, this provides evidence that heroin can increase susceptibility to pathogens by directly interacting with the immune system. Previous research in our laboratory has demonstrated that these immunosuppressive effects of heroin can also be conditioned to environmental stimuli by repeatedly pairing heroin administration with a unique environmental context. Re-exposure to a previously drug-paired context elicits immunosuppressive effects similar to heroin administration alone. In addition, our laboratory has reported that the basolateral amygdala (BLA) and medial nucleus accumbens shell (mNAcS) are critical neural substrates that mediate this conditioned effect. However, our understanding of the contributing mechanisms within these brain regions is limited. It is known that the cytokine interleukin-1 (IL-1) plays an important role in learning and memory. In fact, our laboratory has demonstrated that inhibition of IL-1β expression in the dorsal hippocampus (DH) prior to re-exposure to a heroin-paired context prevents the suppression of measures of NO production. Therefore, the present studies sought to further investigate the role of IL-1 in heroin-conditioned immunosuppression. Blockade of IL-1 signaling in the BLA, but not in the caudate putamen or mNAcS, using IL-1 receptor antagonist (IL-1Ra) attenuated heroin-conditioned immunosuppression of NO production as measured by plasma nitrate/nitrite and iNOS mRNA expression in spleen tissue. Taken together, these findings suggest that IL-1 signaling in the BLA is necessary for the expression of heroin-conditioned immunosuppression of NO production and may be a target for interventions that normalize immune function in heroin users and patient populations exposed to opiate regimens.
Collapse
Affiliation(s)
- Lee W Hutson
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Christina L Lebonville
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Meghan E Jones
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Rita A Fuchs
- Washington State University, College of Veterinary Medicine, Department of Integrative Physiology and Neuroscience, Pullman, WA, USA
| | - Donald T Lysle
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA.
| |
Collapse
|
24
|
Jin Y, Wang R, Yang S, Zhang X, Dai J. Role of Microglia Autophagy in Microglia Activation After Traumatic Brain Injury. World Neurosurg 2017; 100:351-360. [PMID: 28108422 DOI: 10.1016/j.wneu.2017.01.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/09/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVE We evaluated the role of microglia autophagy in microglia activation after traumatic brain injury (TBI) in rats. METHODS TBI was induced by a fluid percussion TBI device. All rats were killed 24 hours after TBI. The ipsilateral hippocampus in all rats was analyzed with hematoxylin-eosin staining. Immunohistochemistry and Western blotting of ionized calcium-binding adapter molecule 1 was used to determine changes in microglia activation. Double staining of microtubule-associated protein light chain 3, Beclin-1, and ionized calcium-binding adapter molecule 1 was used to assess changes of microglia autophagy. Enzyme-linked immunosorbent assay of tumor necrosis factor-α and interleukin-1β was used to evaluate changes in inflammatory responses. Terminal deoxyribonucleotidyl transferase-mediated deoxyuridine 5'-triphosphate nick-end labeling staining was used to determine cell death in the ipsilateral hippocampus. RESULTS At 24 hours after TBI, microglial cells became activated, and the autophagy inhibitor 3-methyladenine (3-MA) further promoted microglia activation. Protein light chain 3- and Beclin-1-positive microglial cells were increased after TBI, whereas 3-MA decreased the number of positive microglial cells, increasing the expression of tumor necrosis factor-α and interleukin-1β; terminal deoxyribonucleotidyl transferase-mediated deoxyuridine 5'-triphosphate nick-end labeling staining demonstrated that 3-MA could increase the number of terminal deoxyribonucleotidyl transferase-mediated deoxyuridine 5'-triphosphate nick-end labeling-positive cells (16.83 ± 0.83 vs. 11 ± 0.82, P < 0.001). CONCLUSIONS Our data demonstrated that TBI induced microglia activation and microglia autophagy. Inhibition of microglia autophagy with 3-MA increased microglia activation and neural apoptosis. These findings indicate that targeting microglia autophagy may be a therapeutic strategy for TBI.
Collapse
Affiliation(s)
- Yichao Jin
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Ran Wang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Shaofeng Yang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China.
| | - Jiong Dai
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Krityakiarana W, Sompup K, Jongkamonwiwat N, Mukda S, Pinilla FG, Govitrapong P, Phansuwan-Pujito P. Effects of melatonin on severe crush spinal cord injury-induced reactive astrocyte and scar formation. J Neurosci Res 2016; 94:1451-1459. [PMID: 27717042 DOI: 10.1002/jnr.23930] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023]
Abstract
The present work aimed at analyzing the effects of melatonin on scar formation after spinal cord injury (SCI). Upregulation of reactive astrocyte under SCI pathological conditions has been presented in several studies. It has been proved that the crucial factor in triggering this upregulation is proinflammatory cytokines. Moreover, scar formation is an important barrier to axonal regeneration through the lesion area. Melatonin plays an important role in reducing inflammation, but its effects on scar formation in the injured spinal cord remain unknown. Hence, we used the model of severe crush injury in mice to investigate the effects of melatonin on scar formation. Mice were randomly separated into four groups; SCI, SCI+Melatonin 1 (single dose), SCI+Melatonin 14 (14 daily doses), and control. Melatonin was administered by intraperitoneal injection (10 mg/kg) after injury. Immunohistochemical analysis, Western blot, and behavioral evaluation were used to explore the effects of melatonin after SCI for 14 days. The melatonin-treated mice presented higher expression of neuronal markers (P < 0.001). Remarkably, the inflammatory response appeared to be greatly reduced in the SCI+Melatonin 14 group (P < 0.001), which also displayed less scar formation (P < 0.05). These findings suggest that melatonin inhibits scar formation by acting on inflammatory cytokines after SCI. Overall, our results suggest that melatonin is a promising treatment strategy after SCI that deserves further investigation. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Warin Krityakiarana
- Division of Physical Therapy, Faculty of Health Science, Srinakharinwirot University, Nakhon-Nayok, Thailand. .,Department of Rehabilitation for Persons with Disabilities, Ratchasuda College, Mahidol University, Nakhonpathom, Thailand.
| | - Kamonrapat Sompup
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand
| | - Nopporn Jongkamonwiwat
- Division of Physical Therapy, Faculty of Health Science, Srinakharinwirot University, Nakhon-Nayok, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand
| | - Fernando Gomez Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand.,Center for Neuroscience and Department for Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
26
|
Sordillo PP, Sordillo LA, Helson L. Bifunctional role of pro-inflammatory cytokines after traumatic brain injury. Brain Inj 2016; 30:1043-53. [DOI: 10.3109/02699052.2016.1163618] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
Guo ML, Liao K, Periyasamy P, Yang L, Cai Y, Callen SE, Buch S. Cocaine-mediated microglial activation involves the ER stress-autophagy axis. Autophagy 2016; 11:995-1009. [PMID: 26043790 DOI: 10.1080/15548627.2015.1052205] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cocaine abuse leads to neuroinflammation, which, in turn, contributes to the pathogenesis of neurodegeneration associated with advanced HIV-1 infection. Autophagy plays important roles in both innate and adaptive immune responses. However, the possible functional link between cocaine and autophagy has not been explored before. Herein, we demonstrate that cocaine exposure induced autophagy in both BV-2 and primary rat microglial cells as demonstrated by a dose- and time-dependent induction of autophagy-signature proteins such as BECN1/Beclin 1, ATG5, and MAP1LC3B. These findings were validated wherein cocaine treatment of BV-2 cells resulted in increased formation of puncta in cells expressing either endogenous MAP1LC3B or overexpressing GFP-MAP1LC3B. Specificity of cocaine-induced autophagy was confirmed by treating cells with inhibitors of autophagy (3-MA and wortmannin). Intriguingly, cocaine-mediated induction of autophagy involved upstream activation of 2 ER stress pathways (EIF2AK3- and ERN1-dependent), as evidenced by the ability of the ER stress inhibitor salubrinal to ameliorate cocaine-induced autophagy. In vivo validation of these findings demonstrated increased expression of BECN1, ATG5, and MAP1LC3B-II proteins in cocaine-treated mouse brains compared to untreated animals. Increased autophagy contributes to cocaine-mediated activation of microglia since pretreatment of cells with wortmannin resulted in decreased expression and release of inflammatory factors (TNF, IL1B, IL6, and CCL2) in microglial cells. Taken together, our findings suggest that cocaine exposure results in induction of autophagy that is closely linked with neuroinflammation. Targeting autophagic proteins could thus be considered as a therapeutic strategy for the treatment of cocaine-related neuroinflammation diseases.
Collapse
Key Words
- 3-MA, 3-methyladenine
- ATF6, activating transcription factor 6
- ATG5, autophagy-related 5
- BCL2, B-cell CLL/lymphoma 2
- BECN1
- BECN1, Beclin 1, autophagy related
- Baf1, bafilomycin A1
- CCL2, chemokine (C-C motif) ligand 2
- DAPI: 4, 6-diamidino-2-phenylindole, dihydrochloride
- DDIT3, DNA-damage-inducible transcript 3
- EGFP, enhanced green fluorescent protein
- EIF2AK3, eukaryotic translation initiation factor 2-α kinase 3
- EIF2S1, eukaryotic translation initiation factor 2, subunit 1 α, 35kDa
- ER stress
- ER, endoplasmic reticulum
- ERN1, endoplasmic reticulum to nucleus signaling 1
- HIV, human immunodeficiency virus
- IL1B, interleukin 1, β
- IL6, interleukin 6
- MAP1LC3B
- MAP1LC3B, microtubule-associated protein 1 light chain 3
- METH, methamphetamine
- MTOR, mechanistic target of rapamycin
- NFKB1, nuclear factor of kappa light polypeptide gene enhancer in B-cells 1
- PBN, N-tert-butyl-α-phenylnitrone
- PPP1R3A, protein phosphatase 1, regulatory subunit 3A
- PtdIns3K, class III phosphatidylinositol 3-kinase
- ROS, reactive oxygen species
- RPS6, ribosomal protein S6
- TLR4, toll-like receptor 4
- TNF, tumor necrosis factor
- autophagy
- cocaine
- microglial cells
- neuroinflammation
- rPMCs, rat primary microglial cells
- wort, wortmannin
Collapse
Affiliation(s)
- Ming-Lei Guo
- a Department of Pharmacology and Experimental Neuroscience; Nebraska Medical Center; University of Nebraska Medical Center ; Omaha , NE , USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Verma AK, Ghosh S, Pradhan S, Basu A. Microglial activation induces neuronal death in Chandipura virus infection. Sci Rep 2016; 6:22544. [PMID: 26931456 PMCID: PMC4773833 DOI: 10.1038/srep22544] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/15/2016] [Indexed: 12/22/2022] Open
Abstract
Neurotropic viruses induce neurodegeneration either directly by activating host death domains or indirectly through host immune response pathways. Chandipura Virus (CHPV) belonging to family Rhabdoviridae is ranked among the emerging pathogens of the Indian subcontinent. Previously we have reported that CHPV induces neurodegeneration albeit the root cause of this degeneration is still an open question. In this study we explored the role of microglia following CHPV infection. Phenotypic analysis of microglia through lectin and Iba-1 staining indicated cells were in an activated state post CHPV infection in cortical region of the infected mouse brain. Cytokine Bead Array (CBA) analysis revealed comparatively higher cytokine and chemokine levels in the same region. Increased level of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), Nitric Oxide (NO) and Reactive Oxygen species (ROS) in CHPV infected mouse brain indicated a strong inflammatory response to CHPV infection. Hence it was hypothesized through our analyses that this inflammatory response may stimulate the neuronal death following CHPV infection. In order to validate our hypothesis supernatant from CHPV infected microglial culture was used to infect neuronal cell line and primary neurons. This study confirmed the bystander killing of neurons due to activation of microglia post CHPV infection.
Collapse
Affiliation(s)
| | - Sourish Ghosh
- National Brain Research Centre, Manesar, Haryana-122051, India
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana-122051, India
| |
Collapse
|
29
|
Swaroop S, Sengupta N, Suryawanshi AR, Adlakha YK, Basu A. HSP60 plays a regulatory role in IL-1β-induced microglial inflammation via TLR4-p38 MAPK axis. J Neuroinflammation 2016; 13:27. [PMID: 26838598 PMCID: PMC4736186 DOI: 10.1186/s12974-016-0486-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/19/2016] [Indexed: 01/08/2023] Open
Abstract
Background IL-1β, also known as “the master regulator of inflammation”, is a potent pro-inflammatory cytokine secreted by activated microglia in response to pathogenic invasions or neurodegeneration. It initiates a vicious cycle of inflammation and orchestrates various molecular mechanisms involved in neuroinflammation. The role of IL-1β has been extensively studied in neurodegenerative disorders; however, molecular mechanisms underlying inflammation induced by IL-1β are still poorly understood. The objective of our study is the comprehensive identification of molecular circuitry involved in IL-1β-induced inflammation in microglia through protein profiling. Methods To achieve our aim, we performed the proteomic analysis of N9 microglial cells with and without IL-1β treatment at different time points. Expression of HSP60 in response to IL-1β administration was checked by quantitative real-time PCR, immunoblotting, and immunofluorescence. Interaction of HSP60 with TLR4 was determined by co-immunoprecipitation. Inhibition of TLR4 was done using TLR4 inhibitor to reveal its effect on IL-1β-induced inflammation. Further, effect of HSP60 knockdown and overexpression were assessed on the inflammation in microglia. Specific MAPK inhibitors were used to reveal the downstream MAPK exclusively involved in HSP60-induced inflammation in microglia. Results Total 21 proteins were found to be differentially expressed in response to IL-1β treatment in N9 microglial cells. In silico analysis of these proteins revealed unfolded protein response as one of the most significant molecular functions, and HSP60 turned out to be a key hub molecule. IL-1β induced the expression as well as secretion of HSP60 in extracellular milieu during inflammation of N9 cells. Secreted HSP60 binds to TLR4 and inhibition of TLR4 suppressed IL-1β-induced inflammation to a significant extent. Our knockdown and overexpression studies demonstrated that HSP60 increases the phosphorylation of ERK, JNK, and p38 MAPKs in N9 cells during inflammation. Specific inhibition of p38 by inhibitors suppressed HSP60-induced inflammation, thus pointed towards the major role of p38 MAPK rather than ERK1/2 and JNK in HSP60-induced inflammation. Furthermore, silencing of upstream modulator of p38, i.e., MEK3/6 also reduced HSP60-induced inflammation. Conclusions IL-1β induces expression of HSP60 in N9 microglial cells that further augments inflammation via TLR4-p38 MAPK axis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0486-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shalini Swaroop
- National Brain Research Centre, Manesar, Haryana, 122051, India
| | | | | | - Yogita K Adlakha
- National Brain Research Centre, Manesar, Haryana, 122051, India.
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, 122051, India.
| |
Collapse
|
30
|
Anrather J, Iadecola C, Hallenbeck J. Inflammation and Immune Response. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
31
|
Sapin E, Peyron C, Roche F, Gay N, Carcenac C, Savasta M, Levy P, Dematteis M. Chronic Intermittent Hypoxia Induces Chronic Low-Grade Neuroinflammation in the Dorsal Hippocampus of Mice. Sleep 2015; 38:1537-46. [PMID: 26085297 DOI: 10.5665/sleep.5042] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Indexed: 12/21/2022] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea (OSA) induces cognitive impairment that involves intermittent hypoxia (IH). Because OSA is recognized as a low-grade systemic inflammatory disease and only some patients develop cognitive deficits, we investigated whether IH-related brain consequences shared similar pathophysiology and required additional factors such as systemic inflammation to develop. DESIGN Nine-week-old male C57BL/6J mice were exposed to 1 day, 6 or 24 w of IH (alternating 21-5% FiO2 every 30 sec, 8 h/day) or normoxia. Microglial changes were assessed in the functionally distinct dorsal (dH) and ventral (vH) regions of the hippocampus using Iba1 immunolabeling. Then the study concerned dH, as vH only tended to be lately affected. Seven proinflammatory and anti-inflammatory cytokine messenger RNA (mRNA) were assessed at all time points using semiquantitative real-time reverse transcription polymerase chain reaction (RT-PCR). Similar mRNA analysis was performed after 6 w IH or normoxia associated for the past 3 w with repeated intraperitoneal low-dose lipopolysaccharide or saline. MEASUREMENTS AND RESULTS Chronic (6, 24 w) but not acute IH induced significant microglial changes in dH only, including increased density and morphological features of microglia priming. In dH, acute but not chronic IH increased IL-1β and RANTES/CCL5 mRNA, whereas the other cytokines remained unchanged. In contrast, chronic IH plus lipopolysaccharide increased interleukin (IL)-6 and IL10 mRNA whereas lipopolysaccharide alone did not affect these cytokines. CONCLUSION The obstructive sleep apnea component intermittent hypoxia (IH) causes low-grade neuroinflammation in the dorsal hippocampus of mice, including early but transient cytokine elevations, delayed but long-term microglial changes, and cytokine response alterations to lipopolysaccharide inflammatory challenge. These changes may contribute to IH-induced cognitive impairment and pathological brain aging.
Collapse
Affiliation(s)
- Emilie Sapin
- Université Grenoble Alpes, Grenoble, F-38042, France.,INSERM U1042, Laboratoire HP2, Grenoble, F-38042, France
| | - Christelle Peyron
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Team SLEEP, F-69372, France.,Université Claude Bernard Lyon 1, Lyon, F-69372, France
| | - Frédéric Roche
- CHU, Hôpital Nord, Service de Physiologie Clinique et de l'Exercice, Saint-Etienne, F-42270, France.,Université Jean Monnet, Saint-Etienne, F-42023, France
| | - Nadine Gay
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Team SLEEP, F-69372, France.,Université Claude Bernard Lyon 1, Lyon, F-69372, France
| | - Carole Carcenac
- Université Grenoble Alpes, Grenoble, F-38042, France.,INSERM U836, Grenoble Institut des Neurosciences, équipe 10, Grenoble, F-38042, France
| | - Marc Savasta
- Université Grenoble Alpes, Grenoble, F-38042, France.,INSERM U836, Grenoble Institut des Neurosciences, équipe 10, Grenoble, F-38042, France
| | - Patrick Levy
- Université Grenoble Alpes, Grenoble, F-38042, France.,INSERM U1042, Laboratoire HP2, Grenoble, F-38042, France.,CHU, Hôpital Michallon, Laboratoires du Sommeil et EFCR, Grenoble F-38043, France
| | - Maurice Dematteis
- Université Grenoble Alpes, Grenoble, F-38042, France.,INSERM U1042, Laboratoire HP2, Grenoble, F-38042, France.,CHU, Hôpital Michallon, Addictologie, Pôle Pluridisciplinaire de Médecine, Grenoble F-38043, France
| |
Collapse
|
32
|
Bruttger J, Karram K, Wörtge S, Regen T, Marini F, Hoppmann N, Klein M, Blank T, Yona S, Wolf Y, Mack M, Pinteaux E, Müller W, Zipp F, Binder H, Bopp T, Prinz M, Jung S, Waisman A. Genetic Cell Ablation Reveals Clusters of Local Self-Renewing Microglia in the Mammalian Central Nervous System. Immunity 2015; 43:92-106. [PMID: 26163371 DOI: 10.1016/j.immuni.2015.06.012] [Citation(s) in RCA: 465] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 03/05/2015] [Accepted: 04/22/2015] [Indexed: 12/23/2022]
Abstract
During early embryogenesis, microglia arise from yolk sac progenitors that populate the developing central nervous system (CNS), but how the tissue-resident macrophages are maintained throughout the organism's lifespan still remains unclear. Here, we describe a system that allows specific, conditional ablation of microglia in adult mice. We found that the microglial compartment was reconstituted within 1 week of depletion. Microglia repopulation relied on CNS-resident cells, independent from bone-marrow-derived precursors. During repopulation, microglia formed clusters of highly proliferative cells that migrated apart once steady state was achieved. Proliferating microglia expressed high amounts of the interleukin-1 receptor (IL-1R), and treatment with an IL-1R antagonist during the repopulation phase impaired microglia proliferation. Hence, microglia have the potential for efficient self-renewal without the contribution of peripheral myeloid cells, and IL-1R signaling participates in this restorative proliferation process.
Collapse
Affiliation(s)
- Julia Bruttger
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany
| | - Simone Wörtge
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany
| | - Federico Marini
- Institute for Medical Biometry, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 69, 55131 Mainz, Germany
| | - Nicola Hoppmann
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr.1, 55131 Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr.1, 55131 Mainz, Germany
| | - Thomas Blank
- Department of Neuropathology & BIOSS Centre for Biological Signaling Studies, University of Freiburg, Breisacher Str. 64, 79098 Freiburg, Germany
| | - Simon Yona
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yochai Wolf
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Matthias Mack
- Department for Internal Medicine, University Hospital, 93042 Regensburg, Germany
| | - Emmanuel Pinteaux
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Frauke Zipp
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr.1, 55131 Mainz, Germany
| | - Harald Binder
- Institute for Medical Biometry, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 69, 55131 Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr.1, 55131 Mainz, Germany
| | - Marco Prinz
- Department of Neuropathology & BIOSS Centre for Biological Signaling Studies, University of Freiburg, Breisacher Str. 64, 79098 Freiburg, Germany
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany.
| |
Collapse
|
33
|
Addington CP, Roussas A, Dutta D, Stabenfeldt SE. Endogenous repair signaling after brain injury and complementary bioengineering approaches to enhance neural regeneration. Biomark Insights 2015; 10:43-60. [PMID: 25983552 PMCID: PMC4429653 DOI: 10.4137/bmi.s20062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) affects 5.3 million Americans annually. Despite the many long-term deficits associated with TBI, there currently are no clinically available therapies that directly address the underlying pathologies contributing to these deficits. Preclinical studies have investigated various therapeutic approaches for TBI: two such approaches are stem cell transplantation and delivery of bioactive factors to mitigate the biochemical insult affiliated with TBI. However, success with either of these approaches has been limited largely due to the complexity of the injury microenvironment. As such, this review outlines the many factors of the injury microenvironment that mediate endogenous neural regeneration after TBI and the corresponding bioengineering approaches that harness these inherent signaling mechanisms to further amplify regenerative efforts.
Collapse
Affiliation(s)
- Caroline P Addington
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Adam Roussas
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Dipankar Dutta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
34
|
Castro-Torres RD, Chaparro-Huerta V, Flores-Soto ME, Jave-Suárez L, Camins A, Armendáriz-Borunda J, Beas-Zárate C, Mena-Munguía S. Pirfenidone Attenuates Microglial Reactivity and Reduces Inducible Nitric Oxide Synthase mRNA Expression After Kainic Acid-Mediated Excitotoxicity in Pubescent Rat Hippocampus. J Mol Neurosci 2015; 56:245-54. [DOI: 10.1007/s12031-015-0509-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/01/2015] [Indexed: 12/20/2022]
|
35
|
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Despite extensive preclinical research supporting the effectiveness of neuroprotective therapies for brain trauma, there have been no successful randomized controlled clinical trials to date. TBI results in delayed secondary tissue injury due to neurochemical, metabolic and cellular changes; modulating such effects has provided the basis for neuroprotective interventions. To establish more effective neuroprotective treatments for TBI it is essential to better understand the complex cellular and molecular events that contribute to secondary injury. Here we critically review relevant research related to causes and modulation of delayed tissue damage, with particular emphasis on cell death mechanisms and post-traumatic neuroinflammation. We discuss the concept of utilizing multipotential drugs that target multiple secondary injury pathways, rather than more specific "laser"-targeted strategies that have uniformly failed in clinical trials. Moreover, we assess data supporting use of neuroprotective drugs that are currently being evaluated in human clinical trials for TBI, as well as promising emerging experimental multipotential drug treatment strategies. Finally, we describe key challenges and provide suggestions to improve the likelihood of successful clinical translation.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
36
|
Bastien D, Lacroix S. Cytokine pathways regulating glial and leukocyte function after spinal cord and peripheral nerve injury. Exp Neurol 2014; 258:62-77. [PMID: 25017888 DOI: 10.1016/j.expneurol.2014.04.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 02/20/2014] [Accepted: 04/08/2014] [Indexed: 01/13/2023]
Abstract
Injury to the nervous system causes the almost immediate release of cytokines by glial cells and neurons. These cytokines orchestrate a complex array of responses leading to microgliosis, immune cell recruitment, astrogliosis, scarring, and the clearance of cellular debris, all steps that affect neuronal survival and repair. This review will focus on cytokines released after spinal cord and peripheral nerve injury and the primary signalling pathways triggered by these inflammatory mediators. Notably, the following cytokine families will be covered: IL-1, TNF, IL-6-like, TGF-β, and IL-10. Whether interfering with cytokine signalling could lead to novel therapies will also be discussed. Finally, the review will address whether manipulating the above-mentioned cytokine families and signalling pathways could exert distinct effects in the injured spinal cord versus peripheral nerve.
Collapse
Affiliation(s)
- Dominic Bastien
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Steve Lacroix
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada..
| |
Collapse
|
37
|
Tarassishin L, Casper D, Lee SC. Aberrant expression of interleukin-1β and inflammasome activation in human malignant gliomas. PLoS One 2014; 9:e103432. [PMID: 25054228 PMCID: PMC4108401 DOI: 10.1371/journal.pone.0103432] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/02/2014] [Indexed: 11/18/2022] Open
Abstract
Objective Glioblastoma is the most frequent and malignant form of primary brain tumor with grave prognosis. Mounting evidence supports that chronic inflammation (such as chronic overactivation of IL-1 system) is a crucial event in carcinogenesis and tumor progression. IL-1 also is an important cytokine with species-dependent regulations and roles in CNS cell activation. While much attention is paid to specific anti-tumor immunity, little is known about the role of chronic inflammation/innate immunity in glioma pathogenesis. In this study, we examined whether human astrocytic cells (including malignant gliomas) can produce IL-1 and its role in glioma progression. Methods We used a combination of cell culture, real-time PCR, ELISA, western blot, immunocytochemistry, siRNA and plasmid transfection, micro-RNA analysis, angiogenesis (tube formation) assay, and neurotoxicity assay. Results Glioblastoma cells produced large quantities of IL-1 when activated, resembling macrophages/microglia. The activation signal was provided by IL-1 but not the pathogenic components LPS or poly IC. Glioblastoma cells were highly sensitive to IL-1 stimulation, suggesting its relevance in vivo. In human astrocytes, IL-1β mRNA was not translated to protein. Plasmid transfection also failed to produce IL-1 protein, suggesting active repression. Suppression of microRNAs that can target IL-1α/β did not induce IL-1 protein. Glioblastoma IL-1β processing occurred by the NLRP3 inflammasome, and ATP and nigericin increased IL-1β processing by upregulating NLRP3 expression, similar to macrophages. RNAi of annexin A2, a protein strongly implicated in glioma progression, prevented IL-1 induction, demonstrating its new role in innate immune activation. IL-1 also activated Stat3, a transcription factor crucial in glioma progression. IL-1 activated glioblastoma-conditioned media enhanced angiogenesis and neurotoxicity. Conclusions Our results demonstrate unique, species-dependent immune activation mechanisms involving human astrocytes and astrogliomas. Specifically, the ability to produce IL-1 by glioblastoma cells may confer them a mesenchymal phenotype including increased migratory capacity, unique gene signature and proinflammatory signaling.
Collapse
Affiliation(s)
- Leonid Tarassishin
- Department of Pathology (Neuropathology), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Diana Casper
- Department of Neurosurgery, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Sunhee C Lee
- Department of Pathology (Neuropathology), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| |
Collapse
|
38
|
Walsh S, Gavin A, Wyatt S, O'Connor C, Keeshan K, Nolan YM, O'Keeffe GW, Sullivan AM. Knockdown of interleukin-1 receptor 1 is not neuroprotective in the 6-hydroxydopamine striatal lesion rat model of Parkinson's disease. Int J Neurosci 2014; 125:70-7. [PMID: 24628580 DOI: 10.3109/00207454.2014.904304] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
It is well established that neuroinflammation is associated with the progression of many neurodegenerative diseases, including Parkinson's disease (PD). Activated microglia and elevated levels of pro-inflammatory cytokines such as interleukin-1β (IL-1β) have been found in the brain and cerebrospinal fluid of PD patients, suggesting that IL-1β may be involved in the pathogenesis of this disease. This study aimed to knock down the expression of the interleukin-1 type 1 receptor (IL-1R1) to evaluate any potential therapeutic effect of limiting the action of IL-1β in the substantia nigra following a unilateral intrastriatal 6-hydroxydopamine (6-OHDA) lesion in rats. Adult Sprague-Dawley rats received intranigral injections of shRNA specific for IL-1R1, followed 2 weeks later by intrastriatal 6-OHDA. Injection of IL-1R1 shRNA did not prevent 6-OHDA-induced loss of motor function or loss of nigral dopamine neurons. IL-1R1 expression was increased in the midbrain following 6-OHDA injection; this effect was attenuated in 6-OHDA-treated animals that had received IL-1R1 shRNA. These data suggest that while IL-1R1 was increased in 6-OHDA-treated animals and reduced following shRNA injection, the neurodegeneration induced by 6-OHDA was not mediated through IL-1R1.
Collapse
Affiliation(s)
- Sinéad Walsh
- 1Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork , Cork , Ireland
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Tarassishin L, Lim J, Weatherly DB, Angeletti RH, Lee SC. Interleukin-1-induced changes in the glioblastoma secretome suggest its role in tumor progression. J Proteomics 2014; 99:152-168. [PMID: 24503185 DOI: 10.1016/j.jprot.2014.01.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 01/02/2014] [Accepted: 01/23/2014] [Indexed: 12/26/2022]
Abstract
UNLABELLED The tumor microenvironment including glial cells and their inflammatory products regulates brain tumor development and progression. We have previously established that human glioma cells are exquisitely sensitive to IL-1 stimulation leading us to undertake a comparative analysis of the secretome of unstimulated and cytokine (IL-1)-stimulated glioblastoma cells. We performed label-free quantitative proteomic analysis and detected 190 proteins which included cytokines, chemokines, growth factors, proteases, cell adhesion molecules, extracellular matrix (ECM) and related proteins. Measuring area under the curve (AUC) of peptides for quantitation, the IL-1-induced secretome contained 13 upregulated and 5 downregulated extracellular proteins (p<0.05) compared to controls. Of these, IL-8, CCL2, TNC, Gal-1 and PTX3 were validated as upregulated and SERPINE1, STC2, CTGF and COL4A2 were validated as downregulated factors by immunochemical methods. A major representation of the ECM and related proteins in the glioblastoma secretome and their modulation by IL-1 suggested that IL-1 induces its effect in part by altering TGFβ expression, activity and signaling. These findings enhance our understanding of IL-1-induced modulation of glioma microenvironment, with implications for increased tumor invasion, migration and angiogenesis. They further provide novel targets for the glioblastoma intervention. BIOLOGICAL SIGNIFICANCE Present study is on an unbiased screening of the glioblastoma secretome stimulated by IL-1 which triggers neuroinflammatory cascades in the central nervous system. Network of secreted proteins were shown to be regulated revealing their possible contribution to glioma progression. Label free quantitative proteomics has provided unique novel targets for potential glioblastoma intervention.
Collapse
Affiliation(s)
- Leonid Tarassishin
- Department of Pathology, Albert Einstein College of Medicine, Bronx NY 10461
| | - Jihyeon Lim
- Department of Pathology, Albert Einstein College of Medicine, Bronx NY 10461.,Laboratory for Macromolecular Analysis & Proteomics, Albert Einstein College of Medicine, Bronx NY 10461
| | - D Brent Weatherly
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602
| | - Ruth H Angeletti
- Laboratory for Macromolecular Analysis & Proteomics, Albert Einstein College of Medicine, Bronx NY 10461.,Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx NY 10461
| | - Sunhee C Lee
- Department of Pathology, Albert Einstein College of Medicine, Bronx NY 10461
| |
Collapse
|
40
|
Erythropoietin improves motor and cognitive deficit, axonal pathology, and neuroinflammation in a combined model of diffuse traumatic brain injury and hypoxia, in association with upregulation of the erythropoietin receptor. J Neuroinflammation 2013; 10:156. [PMID: 24344874 PMCID: PMC3896698 DOI: 10.1186/1742-2094-10-156] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/05/2013] [Indexed: 01/12/2023] Open
Abstract
Background Diffuse axonal injury is a common consequence of traumatic brain injury (TBI) and often co-occurs with hypoxia, resulting in poor neurological outcome for which there is no current therapy. Here, we investigate the ability of the multifunctional compound erythropoietin (EPO) to provide neuroprotection when administered to rats after diffuse TBI alone or with post-traumatic hypoxia. Methods Sprague–Dawley rats were subjected to diffuse traumatic axonal injury (TAI) followed by 30 minutes of hypoxic (Hx, 12% O2) or normoxic ventilation, and were administered recombinant human EPO-α (5000 IU/kg) or saline at 1 and 24 hours post-injury. The parameters examined included: 1) behavioural and cognitive deficit using the Rotarod, open field and novel object recognition tests; 2) axonal pathology (NF-200); 3) callosal degradation (hematoxylin and eosin stain); 3) dendritic loss (MAP2); 4) expression and localisation of the EPO receptor (EpoR); 5) activation/infiltration of microglia/macrophages (CD68) and production of IL-1β. Results EPO significantly improved sensorimotor and cognitive recovery when administered to TAI rats with hypoxia (TAI + Hx). A single dose of EPO at 1 hour reduced axonal damage in the white matter of TAI + Hx rats at 1 day by 60% compared to vehicle. MAP2 was decreased in the lateral septal nucleus of TAI + Hx rats; however, EPO prevented this loss, and maintained MAP2 density over time. EPO administration elicited an early enhanced expression of EpoR 1 day after TAI + Hx compared with a 7-day peak in vehicle controls. Furthermore, EPO reduced IL-1β to sham levels 2 hours after TAI + Hx, concomitant to a decrease in CD68 positive cells at 7 and 14 days. Conclusions When administered EPO, TAI + Hx rats had improved behavioural and cognitive performance, attenuated white matter damage, resolution of neuronal damage spanning from the axon to the dendrite, and suppressed neuroinflammation, alongside enhanced expression of EpoR. These data provide compelling evidence of EPO’s neuroprotective capability. Few benefits were observed when EPO was administered to TAI rats without hypoxia, indicating that EPO’s neuroprotective capacity is bolstered under hypoxic conditions, which may be an important consideration when EPO is employed for neuroprotection in the clinic.
Collapse
|
41
|
McGuire SO, Sortwell CE, Shukitt-Hale B, Joseph JA, Hejna MJ, Collier TJ. Dietary supplementation with blueberry extract improves survival of transplanted dopamine neurons. Nutr Neurosci 2013; 9:251-8. [PMID: 17263092 DOI: 10.1080/10284150601086134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The exact mechanisms contributing to poor neuronal survival in cell transplantation paradigms for Parkinson's disease (PD) are unknown. However, transplantation-induced host immune response, inflammation, and subsequent oxidative stress are likely contributors to cell death since dopamine (DA) neurons are exquisitely sensitive to oxidative damage. Multiple studies have attempted to improve cell survival by treating transplant material with antioxidant and antiinflammatory compounds, whereas far fewer studies have attempted to modify the host environment to reduce these threats. Flavonoids, phytochemicals found in fruits and vegetables, have antioxidant, antiinflammatory, and immunomodulatory properties. For example, supplementation with dietary blueberry extract (BBE) prevents oxidative stress-associated impairment of striatal motor function during aging and restores lost motor function in aged rats. We hypothesized that dietary supplementation of rodent diets with BBE would improve the survival of embryonic DA neurons transplanted into the unilaterally DA-depleted striatum. Inclusion of 2% BBE in a custom chow diet significantly increased the survival of implanted DA neurons and ameliorated rotational behavior asymmetries as compared to transplanted animals consuming a standard diet. These findings provide support for the potential of dietary phytochemicals as an easily administered and well-tolerated therapy that can be used to improve the effectiveness of DA neuron replacement.
Collapse
Affiliation(s)
- Susan O McGuire
- Department of Pathology, Loyola University Medical School, Loyola University Chicago, Maywood, IL 60153, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Woodcock T, Morganti-Kossmann MC. The role of markers of inflammation in traumatic brain injury. Front Neurol 2013; 4:18. [PMID: 23459929 DOI: 10.3389/fneur.2013.00018.ecollection2013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 02/10/2013] [Indexed: 05/19/2023] Open
Abstract
Within minutes of a traumatic impact, a robust inflammatory response is elicited in the injured brain. The complexity of this post-traumatic squeal involves a cellular component, comprising the activation of resident glial cells, microglia, and astrocytes, and the infiltration of blood leukocytes. The second component regards the secretion immune mediators, which can be divided into the following sub-groups: the archetypal pro-inflammatory cytokines (Interleukin-1, Tumor Necrosis Factor, Interleukin-6), the anti-inflammatory cytokines (IL-4, Interleukin-10, and TGF-beta), and the chemotactic cytokines or chemokines, which specifically drive the accumulation of parenchymal and peripheral immune cells in the injured brain region. Such mechanisms have been demonstrated in animal models, mostly in rodents, as well as in human brain. Whilst the humoral immune response is particularly pronounced in the acute phase following Traumatic brain injury (TBI), the activation of glial cells seems to be a rather prolonged effect lasting for several months. The complex interaction of cytokines and cell types installs a network of events, which subsequently intersect with adjacent pathological cascades including oxidative stress, excitotoxicity, or reparative events including angiogenesis, scarring, and neurogenesis. It is well accepted that neuroinflammation is responsible of beneficial and detrimental effects, contributing to secondary brain damage but also facilitating neurorepair. Although such mediators are clear markers of immune activation, to what extent cytokines can be defined as diagnostic factors reflecting brain injury or as predictors of long term outcome needs to be further substantiated. In clinical studies some groups reported a proportional cytokine production in either the cerebrospinal fluid or intraparenchymal tissue with initial brain damage, mortality, or poor outcome scores. However, the validity of cytokines as biomarkers is not broadly accepted. This review article will discuss the evidence from both clinical and laboratory studies exploring the validity of immune markers as a correlate to classification and outcome following TBI.
Collapse
Affiliation(s)
- Thomas Woodcock
- Australian School of Advanced Medicine, Macquarie University Sydney, NSW, Australia
| | | |
Collapse
|
43
|
Hassani Z, O'Reilly J, Pearse Y, Stroemer P, Tang E, Sinden J, Price J, Thuret S. Human neural progenitor cell engraftment increases neurogenesis and microglial recruitment in the brain of rats with stroke. PLoS One 2012. [PMID: 23185625 PMCID: PMC3503964 DOI: 10.1371/journal.pone.0050444] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Main Objectives Stem cell transplantation is to date one of the most promising therapies for chronic ischemic stroke. The human conditionally immortalised neural stem cell line, CTX0E03, has demonstrable efficacy in a rodent model of stroke and is currently in clinical trials. Nonetheless, the mechanisms by which it promotes brain repair are not fully characterised. This study investigated the cellular events occurring after CTX0E03 transplantation in the brains of rats that underwent ischemic stroke. Methods We focused on the endogenous proliferative activity of the host brain in response to cell transplantation and determined the identity of the proliferating cells using markers for young neurons (doublecortin, Dcx) and microglia (CD11b). So as to determine the chronology of events occurring post-transplantation, we analysed the engrafted brains one week and four weeks post-transplantation. Results We observed a significantly greater endogenous proliferation in the striatum of ischemic brains receiving a CTX0E03 graft compared to vehicle-treated ischemic brains. A significant proportion of these proliferative cells were found to be Dcx+ striatal neuroblasts. Further, we describe an enhanced immune response after CTX0E03 engraftment, as shown by a significant increase of proliferating CD11b+ microglial cells. Conclusions Our study demonstrates that few Dcx+ neuroblasts are proliferative in normal conditions, and that this population of proliferative neuroblasts is increased in response to stroke. We further show that CTX0E03 transplantation after stroke leads to the maintenance of this proliferative activity. Interestingly, the preservation of neuronal proliferative activity upon CTX0E03 transplantation is preceded and accompanied by a high rate of proliferating microglia. Our study suggests that microglia might mediate in part the effect of CTX0E03 transplantation on neuronal proliferation in ischemic stroke conditions.
Collapse
Affiliation(s)
- Zahra Hassani
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
| | - Joanna O'Reilly
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
| | - Yewande Pearse
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
| | - Paul Stroemer
- ReNeuron Group plc, Guildford, Surrey, United Kingdom
| | - Ellen Tang
- ReNeuron Group plc, Guildford, Surrey, United Kingdom
| | - John Sinden
- ReNeuron Group plc, Guildford, Surrey, United Kingdom
| | - Jack Price
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
- * E-mail: (JP); (ST)
| | - Sandrine Thuret
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
- * E-mail: (JP); (ST)
| |
Collapse
|
44
|
Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun 2012; 26:1191-201. [PMID: 22728326 DOI: 10.1016/j.bbi.2012.06.008] [Citation(s) in RCA: 498] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/27/2012] [Accepted: 06/14/2012] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) remains one of the leading causes of mortality and morbidity worldwide, yet despite extensive efforts to develop neuroprotective therapies for this devastating disorder there have been no successful outcomes in human clinical trials to date. Following the primary mechanical insult TBI results in delayed secondary injury events due to neurochemical, metabolic and cellular changes that account for many of the neurological deficits observed after TBI. The development of secondary injury represents a window of opportunity for therapeutic intervention to prevent progressive tissue damage and loss of function after injury. To establish effective neuroprotective treatments for TBI it is essential to fully understand the complex cellular and molecular events that contribute to secondary injury. Neuroinflammation is well established as a key secondary injury mechanism after TBI, and it has been long considered to contribute to the damage sustained following brain injury. However, experimental and clinical research indicates that neuroinflammation after TBI can have both detrimental and beneficial effects, and these likely differ in the acute and delayed phases after injury. The key to developing future anti-inflammatory based neuroprotective treatments for TBI is to minimize the detrimental and neurotoxic effects of neuroinflammation while promoting the beneficial and neurotrophic effects, thereby creating optimal conditions for regeneration and repair after injury. This review outlines how post-traumatic neuroinflammation contributes to secondary injury after TBI, and discusses the complex and varied responses of the primary innate immune cells of the brain, microglia, to injury. In addition, emerging experimental anti-inflammatory and multipotential drug treatment strategies for TBI are discussed, as well as some of the challenges faced by the research community to translate promising neuroprotective drug treatments to the clinic.
Collapse
Affiliation(s)
- Alok Kumar
- Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, United States
| | | |
Collapse
|
45
|
Sung CS, Cherng CH, Wen ZH, Chang WK, Huang SY, Lin SL, Chan KH, Wong CS. Minocycline and fluorocitrate suppress spinal nociceptive signaling in intrathecal IL-1β-induced thermal hyperalgesic rats. Glia 2012; 60:2004-17. [PMID: 22972308 DOI: 10.1002/glia.22415] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 08/15/2012] [Indexed: 12/30/2022]
Abstract
We previously demonstrated that intrathecal IL-1β caused thermal hyperalgesia in rats. This study was conducted to examine the effects and cellular mechanisms of glial inhibitors on IL-1β-induced nociception in rats. The effects of minocycline (20 μg), fluorocitrate (1 nmol), and SB203580 (5 μg) on IL-1β (100 ng) treatment in rats were measured by nociceptive behaviors, western blotting of p38 mitogen-activated protein kinase (MAPK) and inducible nitric oxide synthase (iNOS) expression, cerebrospinal fluid nitric oxide (NO) levels, and immunohistochemical analyses. The results demonstrated that intrathecal IL-1β activated microglia and astrocytes, but not neurons, in the dorsal horn of the lumbar spinal cord, as evidenced by morphological changes and increased immunoreactivity, phosphorylated p38 (P-p38) MAPK, and iNOS expression; the activation of microglia and astrocytes peaked at 30 min and lasted for 6 h. The immunoreactivities of microglia and astrocytes were significantly increased at 30 min (6.6- and 2.7-fold, respectively) and 6 h (3.3- and 4.0-fold, respectively) following IL-1β injection, as compared with saline controls at 30 min (all P < 0.01). IL-1β induced P-p38 MAPK and iNOS expression predominantly in microglia and less in astrocytes. Minocycline, fluorocitrate, or SB203580 pretreatment suppressed this IL-1β-upregulated P-p38 MAPK mainly in microglia and iNOS mainly in astrocytes; minocycline exhibited the most potent effect. Minocycline and fluorocitrate pretreatment abrogated IL-1β-induced NO release and thermal hyperalgesia in rats. In conclusion, minocycline, fluorocitrate, and SB203580 effectively suppressed the IL-1β-induced central sensitization and hyperalgesia in rats.
Collapse
Affiliation(s)
- Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Müller MM, Middelanis J, Meier C, Surbek D, Berger R. 17β-estradiol protects 7-day old rats from acute brain injury and reduces the number of apoptotic cells. Reprod Sci 2012; 20:253-61. [PMID: 22875845 DOI: 10.1177/1933719112452471] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To test a possible neuroprotective activity of 17β-estradiol in the neonatal rat brain exposed to hypoxic-ischemia (controlled hypoxia after unilateral carotid artery ligation). METHODS Seven-day-old Wistar rats underwent ligation of the left common carotid artery followed by 80 minutes hypoxia in 8% oxygen inducing an ipsilateral brain damage. Seven days later (d14), brains were analyzed quantitatively using a macroscopic and microscopic score for structural damage, hemisphere volumes were calculated, and immunohistochemistry for cleaved-caspase-3 (marker for apoptotic cells) was performed. Animals from the study group (n = 19) received 17β-estradiol (0.05 µg/g body weight intraperitoneally) before (-64, -40, and -16 hours) and after (+3 hours) the hypoxia (hour 0: start of the hypoxia) and the control group (n = 21) received mock treatment. RESULTS Of the 21 pups, 13 in the NaCl group had macroscopically a severe brain damage and 7 of 19 animals in the study group encountered only discrete to mild lesions. Microscopic brain damage in the study group was significantly lower (score 1.5 ± 0.7 vs 2.8 ± 0.8, P < .05). The determined volumes of the affected hemisphere were significantly lower in the NaCl group than in the treatment group. The numbers of apoptotic cells in both hemispheres was equal in the estradiol group, but in the control group, there were significantly more apoptotic cells in the affected hemisphere (control group: ipsilateral: 1435 ± 653 vs contralateral: 143 ± 57 cells, P < .05). DISCUSSION 17β-Estradiol protects newborn rat brains from hypoxic-ischemic injury, in terms of both microscopic cell injury and apoptosis.
Collapse
Affiliation(s)
- Martin M Müller
- Department of Obstetrics and Gynecology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.
| | | | | | | | | |
Collapse
|
47
|
Ashafaq M, Khan MM, Shadab Raza S, Ahmad A, Khuwaja G, Javed H, Khan A, Islam F, Siddiqui MS, Safhi MM, Islam F. S-allyl cysteine mitigates oxidative damage and improves neurologic deficit in a rat model of focal cerebral ischemia. Nutr Res 2012; 32:133-43. [PMID: 22348462 DOI: 10.1016/j.nutres.2011.12.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/20/2011] [Accepted: 12/30/2011] [Indexed: 01/08/2023]
Abstract
Oxidative stress and inflammatory damage play an important role in cerebral ischemic pathogenesis and may represent a target for treatment. The present study examined the hypothesis that S-allyl cysteine (SAC), organosulfur compounds found in garlic extract, would reduce oxidative stress-associated brain injury after middle cerebral artery occlusion (MCAO). To test this hypothesis, male Wistar rats were subjected to MCAO for 2 hours and 22-hour reperfusion. S-allyl cysteine was administered (100 mg/kg, b.wt.) intraperitoneally 30 minutes before the onset of ischemia and after the ischemia at the interval of 0, 6, and 12 hours. After 24 hours of reperfusion, rats were tested for neurobehavioral activities and were killed for the infarct volume, estimation of lipid peroxidation, glutathione content, and activity of antioxidant enzymes (glutathione peroxidase, glutathione reductase, catalase, and superoxide dismutase). S-allyl cysteine treatment significantly reduced ischemic lesion volume, improved neurologic deficits, combated oxidative loads, and suppressed neuronal loss. Behavioral and biochemical alterations observed after MCAO were further associated with an increase in glial fibrillary acidic protein and inducible nitric oxide expression and were markedly inhibited by the treatment with SAC. The results suggest that SAC exhibits exuberant neuroprotective potential in rat ischemia/reperfusion model. Thus, this finding of SAC-induced adaptation to ischemic stress and inflammation could suggest a novel avenue for clinical intervention during ischemia and reperfusion.
Collapse
Affiliation(s)
- Mohammad Ashafaq
- Department of Medical Elementology and Toxicology (Fund for the Improvement of Science and Technology sponsored by DST and Special Assistance Programme sponsored by UGC), JamiaHamdard (Hamdard University), Hamdard Nagar, New Delhi-110062, India
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Psychostimulant abuse and neuroinflammation: emerging evidence of their interconnection. Neurotox Res 2012; 23:174-88. [PMID: 22714667 DOI: 10.1007/s12640-012-9334-7] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/09/2012] [Accepted: 05/22/2012] [Indexed: 12/31/2022]
Abstract
During the past two decades, there has been a tremendous expansion of knowledge regarding the neurobiological effects of substance abuse and how these effects impact behavior. At the same time, there has been a profound change in our understanding of the way in which the central nervous system responds to noxious stimuli. Most often referred to as the innate immune response (IIR), this defense mechanism is activated by a number of agents (toxic, microbial, ischemic) and has been implicated in the progression of a number of neurodegenerative diseases. We review evidence that psychostimulants of abuse (cocaine, methamphetamine, ecstasy) are associated with activation of the IIR. We first present background on what is currently known about the IIR including some of the cellular elements involved (microglia, astrocytes, vascular endothelial cells), key receptor pathways, and primary inflammatory cytokines (IL-1β, IL-6, TNF-α). We then present a variety of protein and gene expression data taken from animal studies that show increased expression of various components of the IIR following acute or repeated psychostimulant administration. Collectively the data indicate an association of psychostimulant use with IIR activation in the brain even at exposures not traditionally associated with neurotoxicity. Thus, the gradually escalating deleterious effects of psychostimulant use could in part involve neuroinflammatory mechanisms. Finally, we offer one hypothesis of a possible mechanism by which psychostimulants result in IIR activation and discuss the potential therapeutic implications of these findings for treatment of the recovering addict.
Collapse
|
49
|
Guilty molecules, guilty minds? The conflicting roles of the innate immune response to traumatic brain injury. Mediators Inflamm 2012; 2012:356494. [PMID: 22701273 PMCID: PMC3373171 DOI: 10.1155/2012/356494] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/26/2012] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a complex disease in the most complex organ of the body, whose victims endure lifelong debilitating physical, emotional, and psychosocial consequences. Despite advances in clinical care, there is no effective neuroprotective therapy for TBI, with almost every compound showing promise experimentally having disappointing results in the clinic. The complex and highly interrelated innate immune responses govern both the beneficial and deleterious molecular consequences of TBI and are present as an attractive therapeutic target. This paper discusses the positive, negative, and often conflicting roles of the innate immune response to TBI in both an experimental and clinical settings and highlights recent advances in the search for therapeutic candidates for the treatment of TBI.
Collapse
|
50
|
Ferreira R, Santos T, Gonçalves J, Baltazar G, Ferreira L, Agasse F, Bernardino L. Histamine modulates microglia function. J Neuroinflammation 2012; 9:90. [PMID: 22569158 PMCID: PMC3583187 DOI: 10.1186/1742-2094-9-90] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 05/08/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Histamine is commonly acknowledged as an inflammatory mediator in peripheral tissues, leaving its role in brain immune responses scarcely studied. Therefore, our aim was to uncover the cellular and molecular mechanisms elicited by this molecule and its receptors in microglia-induced inflammation by evaluating cell migration and inflammatory mediator release. METHODS Firstly, we detected the expression of all known histamine receptor subtypes (H1R, H2R, H3R and H4R), using a murine microglial cell line and primary microglia cell cultures from rat cortex, by real-time PCR analysis, immunocytochemistry and Western blotting. Then, we evaluated the role of histamine in microglial cell motility by performing scratch wound assays. Results were further confirmed using murine cortex explants. Finally, interleukin-1beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) levels were evaluated by ELISA measurements to determine the role of histamine on the release of these inflammatory mediators. RESULTS After 12 h of treatment, 100 μM histamine and 10 μg/ml histamine-loaded poly (lactic-co-glycolic acid) microparticles significantly stimulated microglia motility via H4R activation. In addition, migration involves α5β1 integrins, and p38 and Akt signaling pathways. Migration of microglial cells was also enhanced in the presence of lipopolysaccharide (LPS, 100 ng/ml), used as a positive control. Importantly, histamine inhibited LPS-stimulated migration via H4R activation. Histamine or H4R agonist also inhibited LPS-induced IL-1β release in both N9 microglia cell line and hippocampal organotypic slice cultures. CONCLUSIONS To our knowledge, we are the first to show a dual role of histamine in the modulation of microglial inflammatory responses. Altogether, our data suggest that histamine per se triggers microglia motility, whereas histamine impedes LPS-induced microglia migration and IL-1β release. This last datum assigns a new putative anti-inflammatory role for histamine, acting via H4R to restrain exacerbated microglial responses under inflammatory challenge, which could have strong repercussions in the treatment of CNS disorders accompanied by microglia-derived inflammation.
Collapse
Affiliation(s)
- Raquel Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | |
Collapse
|