1
|
Jeon E, Seo MS, Lkhagva-Yondon E, Lim YR, Kim SW, Kang YJ, Lee JS, Lee BD, Wi R, Won SY, Chung YC, Park ES, Kim E, Jin BK, Jeon MS. Neuroprotective effect of L-DOPA-induced interleukin-13 on striatonigral degeneration in cerebral ischemia. Cell Death Dis 2024; 15:854. [PMID: 39578419 PMCID: PMC11584695 DOI: 10.1038/s41419-024-07252-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Levodopa (L-DOPA) treatment is a clinically effective strategy for improving motor function in patients with ischemic stroke. However, the mechanisms by which modulating the dopamine system relieves the pathology of the ischemic brain remain unclear. Emerging evidence from an experimental mouse model of ischemic stroke, established by middle cerebral artery occlusion (MCAO), suggested that L-DOPA has the potential to modulate the inflammatory and immune response that occurs during a stroke. Here, we aimed to demonstrate the therapeutic effect of L-DOPA in regulating the systemic immune response and improving functional deficits in mice with ischemia. Transient MCAO led to progressive degeneration of nigrostriatal dopamine neurons and significant rotational behavior in mice. Exogenous L-DOPA treatment attenuated the striatonigral degeneration and reversed motor behavioral impairment. Notably, treatment with L-DOPA significantly increased IL-13 but reduced IFN-γ in infarct lesions. To investigate the role of IL-13 in motor behavior, we stereotaxically injected anti-IL-13 antibodies into the infarct area of the mouse brain one week after MCAO, followed by L-DOPA treatment. The intervention reduced dopamine, IL-13, and IL-10 levels and exacerbated motor function. IL-13 is potentially expressed on CD4 T cells, while IL-10 is mainly expressed on microglia rather than astrocytes. Finally, IL-13 activates the phagocytosis of microglia, which may contribute to neuroprotection by eliminating degenerating neurons. Our study provides evidence that the L-DOPA-activated dopamine system modulates peripheral immune cells, resulting in the expression of anti-inflammatory and neuroprotective cytokines in mice with ischemic stroke.
Collapse
Affiliation(s)
- Eunhae Jeon
- Translational Research Center, Inha University Hospital, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Myeong-Seong Seo
- Translational Research Center, Inha University Hospital, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Enkhmaa Lkhagva-Yondon
- Translational Research Center, Inha University Hospital, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Yu-Ree Lim
- Translational Research Center, Inha University Hospital, Incheon, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Seung-Woo Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Yu Jeong Kang
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jun Seok Lee
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Byoung Dae Lee
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Rayul Wi
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - So-Yoon Won
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Young Cheul Chung
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Predictive Toxicology, Korea Institute of Toxicology 1, Human and Environmental Toxicology, University of Science and Technology, Daejeon, Republic of Korea
| | - Eun S Park
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Byung Kwan Jin
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Myung-Shin Jeon
- Translational Research Center, Inha University Hospital, Incheon, Republic of Korea.
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
2
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
3
|
Pybus AF, Bitarafan S, Brothers RO, Rohrer A, Khaitan A, Moctezuma FR, Udeshi K, Davies B, Triplett S, Griffin MN, Dammer EB, Rangaraju S, Buckley EM, Wood LB. Profiling the neuroimmune cascade in 3xTg-AD mice exposed to successive mild traumatic brain injuries. J Neuroinflammation 2024; 21:156. [PMID: 38872143 PMCID: PMC11177462 DOI: 10.1186/s12974-024-03128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/12/2024] [Indexed: 06/15/2024] Open
Abstract
Repetitive mild traumatic brain injuries (rmTBI) sustained within a window of vulnerability can result in long term cognitive deficits, depression, and eventual neurodegeneration associated with tau pathology, amyloid beta (Aβ) plaques, gliosis, and neuronal and functional loss. However, a comprehensive study relating acute changes in immune signaling and glial reactivity to neuronal changes and pathological markers after single and repetitive mTBIs is currently lacking. In the current study, we addressed the question of how repeated injuries affect the brain neuroimmune response in the acute phase of injury (< 24 h) by exposing the 3xTg-AD mouse model of tau and Aβ pathology to successive (1x-5x) once-daily weight drop closed-head injuries and quantifying immune markers, pathological markers, and transcriptional profiles at 30 min, 4 h, and 24 h after each injury. We used young adult 2-4 month old 3xTg-AD mice to model the effects of rmTBI in the absence of significant tau and Aβ pathology. We identified pronounced sexual dimorphism in this model, with females eliciting more diverse changes after injury compared to males. Specifically, females showed: (1) a single injury caused a decrease in neuron-enriched genes inversely correlated with inflammatory protein expression and an increase in AD-related genes within 24 h, (2) each injury significantly increased a group of cortical cytokines (IL-1α, IL-1β, IL-2, IL-9, IL-13, IL-17, KC) and MAPK phospho-proteins (phospho-Atf2, phospho-Mek1), several of which co-labeled with neurons and correlated with phospho-tau, and (3) repetitive injury caused increased expression of genes associated with astrocyte reactivity and macrophage-associated immune function. Collectively our data suggest that neurons respond to a single injury within 24 h, while other cell types, including astrocytes, transition to inflammatory phenotypes within days of repetitive injury.
Collapse
Affiliation(s)
- Alyssa F Pybus
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sara Bitarafan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rowan O Brothers
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Alivia Rohrer
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Arushi Khaitan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Felix Rivera Moctezuma
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kareena Udeshi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Brae Davies
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sydney Triplett
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Martin N Griffin
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Eric B Dammer
- Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | - Srikant Rangaraju
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Erin M Buckley
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA.
- Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
4
|
Xie Q, Namba MD, Buck LA, Park K, Jackson JG, Barker JM. Effects of Antiretroviral Treatment on Central and Peripheral Immune Response in Mice with EcoHIV Infection. Cells 2024; 13:882. [PMID: 38786105 PMCID: PMC11120433 DOI: 10.3390/cells13100882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
HIV infection is an ongoing global health issue, despite increased access to antiretroviral therapy (ART). People living with HIV (PLWH) who are virally suppressed through ART still experience negative health outcomes, including neurocognitive impairment. It is increasingly evident that ART may act independently or in combination with HIV infection to alter the immune state, though this is difficult to disentangle in the clinical population. Thus, these experiments used multiplexed chemokine/cytokine arrays to assess peripheral (plasma) and brain (nucleus accumbens; NAc) expression of immune targets in the presence and absence of ART treatment in the EcoHIV mouse model. The findings identify the effects of EcoHIV infection and of treatment with bictegravir (B), emtricitabine (F), and tenofovir alafenamide (TAF) on the expression of numerous immune targets. In the NAc, this included EcoHIV-induced increases in IL-1α and IL-13 expression and B/F/TAF-induced reductions in KC/CXCL1. In the periphery, EcoHIV suppressed IL-6 and LIF expression, while B/F/TAF reduced IL-12p40 expression. In the absence of ART, IBA-1 expression was negatively correlated with CX3CL1 expression in the NAc of EcoHIV-infected mice. These findings identify distinct effects of ART and EcoHIV infection on peripheral and central immune factors and emphasize the need to consider ART effects on neural and immune outcomes.
Collapse
Affiliation(s)
- Qiaowei Xie
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Q.X.); (M.D.N.); (L.A.B.); (J.G.J.)
- Graduate Program in Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Mark D. Namba
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Q.X.); (M.D.N.); (L.A.B.); (J.G.J.)
| | - Lauren A. Buck
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Q.X.); (M.D.N.); (L.A.B.); (J.G.J.)
| | - Kyewon Park
- Center for AIDS Research, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Joshua G. Jackson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Q.X.); (M.D.N.); (L.A.B.); (J.G.J.)
| | - Jacqueline M. Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Q.X.); (M.D.N.); (L.A.B.); (J.G.J.)
| |
Collapse
|
5
|
Pybus AF, Bitarafan S, Brothers RO, Rohrer A, Khaitan A, Moctezuma FR, Udeshi K, Davies B, Triplett S, Dammer E, Rangaraju S, Buckley EM, Wood LB. Profiling the neuroimmune cascade in 3xTg mice exposed to successive mild traumatic brain injuries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544838. [PMID: 37397993 PMCID: PMC10312742 DOI: 10.1101/2023.06.13.544838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Repetitive mild traumatic brain injuries (rmTBI) sustained within a window of vulnerability can result in long term cognitive deficits, depression, and eventual neurodegeneration associated with tau pathology, amyloid beta (Aβ) plaques, gliosis, and neuronal and functional loss. However, we have limited understanding of how successive injuries acutely affect the brain to result in these devastating long-term consequences. In the current study, we addressed the question of how repeated injuries affect the brain in the acute phase of injury (<24hr) by exposing the 3xTg-AD mouse model of tau and Aβ pathology to successive (1x, 3x, 5x) once-daily weight drop closed-head injuries and quantifying immune markers, pathological markers, and transcriptional profiles at 30min, 4hr, and 24hr after each injury. We used young adult mice (2-4 months old) to model the effects of rmTBI relevant to young adult athletes, and in the absence of significant tau and Aβ pathology. Importantly, we identified pronounced sexual dimorphism, with females eliciting more differentially expressed proteins after injury compared to males. Specifically, females showed: 1) a single injury caused a decrease in neuron-enriched genes inversely correlated with inflammatory protein expression as well as an increase in AD-related genes within 24hr, 2) each injury significantly increased expression of a group of cortical cytokines (IL-1α, IL-1β, IL-2, IL-9, IL-13, IL-17, KC) and MAPK phospho-proteins (phospho-Atf2, phospho-Mek1), several of which were co-labeled with neurons and correlated with phospho-tau, and 3) repetitive injury caused increased expression of genes associated with astrocyte reactivity and immune function. Collectively our data suggest that neurons respond to a single injury within 24h, while other cell types including astrocytes transition to inflammatory phenotypes within days of repetitive injury.
Collapse
|
6
|
Interleukin-13 and its receptor are synaptic proteins involved in plasticity and neuroprotection. Nat Commun 2023; 14:200. [PMID: 36639371 PMCID: PMC9839781 DOI: 10.1038/s41467-023-35806-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Immune system molecules are expressed by neurons, yet their functions are often unknown. We have identified IL-13 and its receptor IL-13Ra1 as neuronal, synaptic proteins in mouse, rat, and human brains, whose engagement upregulates the phosphorylation of NMDAR and AMPAR subunits and, in turn, increases synaptic activity and CREB-mediated transcription. We demonstrate that increased IL-13 is a hallmark of traumatic brain injury (TBI) in male mice as well as in two distinct cohorts of human patients. We also provide evidence that IL-13 upregulation protects neurons from excitotoxic death. We show IL-13 upregulation occurring in several cohorts of human brain samples and in cerebrospinal fluid (CSF). Thus, IL-13 is a physiological modulator of synaptic physiology of neuronal origin, with implications for the establishment of synaptic plasticity and the survival of neurons under injury conditions. Furthermore, we suggest that the neuroprotection afforded through the upregulation of IL-13 represents an entry point for interventions in the pathophysiology of TBI.
Collapse
|
7
|
Tatu AL, Nadasdy T, Arbune A, Chioncel V, Bobeica C, Niculet E, Iancu AV, Dumitru C, Popa VT, Kluger N, Clatici VG, Vasile CI, Onisor C, Nechifor A. Interrelationship and Sequencing of Interleukins4, 13, 31, and 33 - An Integrated Systematic Review: Dermatological and Multidisciplinary Perspectives. J Inflamm Res 2022; 15:5163-5184. [PMID: 36110506 PMCID: PMC9468867 DOI: 10.2147/jir.s374060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022] Open
Abstract
The interrelations and sequencing of interleukins are complex (inter)actions where each interleukin can stimulate the secretion of its preceding interleukin. In this paper, we attempt to summarize the currently known roles of IL-4, IL-13, IL-31, and IL-33 from a multi-disciplinary perspective. In order to conduct a comprehensive review of the current literature, a search was conducted using PubMed, Google Scholar, Medscape, UpToDate, and Key Elsevier for keywords. The results were compiled from case reports, case series, letters, and literature review papers, and analyzed by a panel of multi-disciplinary specialist physicians for relevance. Based on 173 results, we compiled the following review of interleukin signaling and its clinical significance across a multitude of medical specialties. Interleukins are at the bed rock of a multitude of pathologies across different organ systems and understanding their role will likely lead to novel treatments and better outcomes for our patients. New interleukins are being described, and the role of this inflammatory cascade is still coming to light. We hope this multi-discipline review on the role interleukins play in current pathology assists in this scope.
Collapse
Affiliation(s)
- Alin Laurentiu Tatu
- Dermatology Department, "Sf. Cuvioasa Parascheva" Clinical Hospital of Infectious Diseases, Galati, Romania.,Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania.,Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania
| | - Thomas Nadasdy
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania.,Dermatology Department, Municipal Emergency Hospital, Timişoara, Romania
| | - Anca Arbune
- Neurology Department, Fundeni Clinical Institute, Bucharest, Romania
| | - Valentin Chioncel
- Neurology Department, "Bagdasar-Arseni" Emergency Clinical Hospital, Bucharest, Romania
| | - Carmen Bobeica
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Elena Niculet
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Caterina Dumitru
- Pharmaceutical Sciences Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| | - Valentin Tudor Popa
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania.,Dermatology Department, Center for the Morphologic Study of the Skin MORPHODERM, "Victor Babeș" University of Medicine and Pharmacy, Timișoara, Romania
| | - Nicolas Kluger
- Department of Dermatology, Allergology and Venereology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.,Apolo Medical Center, Bucharest, Romania
| | | | - Claudiu Ionut Vasile
- Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| | - Cristian Onisor
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Alexandru Nechifor
- Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| |
Collapse
|
8
|
Hong AR, Jang JG, Chung YC, Won SY, Jin BK. Interleukin 13 on Microglia is Neurotoxic in Lipopolysaccharide-injected Striatum in vivo. Exp Neurobiol 2022; 31:42-53. [PMID: 35256543 PMCID: PMC8907255 DOI: 10.5607/en21032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/19/2022] Open
Abstract
To explore the potential function of interleukin-13 (IL-13), lipopolysaccharide (LPS) or PBS as a control was unilaterally microinjected into striatum of rat brain. Seven days after LPS injection, there was a significant loss of neurons and microglial activation in the striatum, visualized by immunohistochemical staining against neuronal nuclei (NeuN) and the OX-42 (complement receptor type 3, CR3), respectively. In parallel, IL-13 immunoreactivity was increased as early as 3 days and sustained up to 7 days post LPS injection, compared to PBS-injected control and detected exclusively within microglia. Moreover, GFAP immunostaining and blood brain barrier (BBB) permeability evaluation showed the loss of astrocytes and disruption of BBB, respectively. By contrast, treatment with IL-13 neutralizing antibody (IL-13NA) protects NeuN+ neurons against LPS-induced neurotoxicity in vivo . Accompanying neuroprotection, IL-13NA reduced loss of GFAP+ astrocytes and damage of BBB in LPS-injected striatum. Intriguingly, treatment with IL-13NA produced neurotrophic factors (NTFs) on survived astrocytes in LPS-injected rat striatum. Taken together, the present study suggests that LPS induces expression of IL-13 on microglia, which contributes to neurodegeneration via damage on astrocytes and BBB disruption in the striatum in vivo.
Collapse
Affiliation(s)
- Ah Reum Hong
- Department of Neuroscience, Graduate School, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jae Geun Jang
- Department of Neuroscience, Graduate School, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Young Cheul Chung
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - So-Yoon Won
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Byung Kwan Jin
- Department of Neuroscience, Graduate School, School of Medicine, Kyung Hee University, Seoul 02447, Korea.,Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
9
|
Barut J, Rafa-Zabłocka K, Jurga AM, Bagińska M, Nalepa I, Parlato R, Kreiner G. Genetic lesions of the noradrenergic system trigger induction of oxidative stress and inflammation in the ventral midbrain. Neurochem Int 2022; 155:105302. [PMID: 35150790 DOI: 10.1016/j.neuint.2022.105302] [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: 08/04/2021] [Revised: 01/07/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits caused by the loss of dopaminergic neurons in the substantia nigra (SN) and ventral tegmental area (VTA). However, clinical data revealed that not only the dopaminergic system is affected in PD. Postmortem studies showed degeneration of noradrenergic cells in the locus coeruleus (LC) to an even greater extent than that observed in the SN/VTA. Pharmacological models support the concept that modification of noradrenergic transmission can influence the PD-like phenotype induced by neurotoxins. Nevertheless, there are no existing data on animal models regarding the distant impact of noradrenergic degeneration on intact SN/VTA neurons. The aim of this study was to create a transgenic mouse model with endogenously evoked progressive degeneration restricted to noradrenergic neurons and investigate its long-term impact on the dopaminergic system. To this end, we selectively ablated the transcription initiation factor-IA (TIF-IA) in neurons expressing dopamine β-hydroxylase (DBH) by the Cre-loxP system. This mutation mimics a condition of nucleolar stress affecting neuronal survival. TIF-IADbhCre mice were characterized by selective, progressive degeneration of noradrenergic neurons, followed by phenotypic alterations associated with sympathetic system impairment. Our studies did not show any loss of tyrosine hydroxylase (TH)-positive cells in the SN/VTA of mutant mice; however, we observed increased indices of oxidative stress, enhanced markers of glial cell activation, inflammatory processes and isolated degenerating cells positive for FluoroJade C. These results were supported by gene expression profiling of VTA and SN from TIF-IADbhCre mice, revealing that 34 out of 246 significantly regulated genes in the SN/VTA were related to PD. Overall, our results shed new light on the possible negative influence of noradrenergic degeneration on dopaminergic neurons, reinforcing the neuroprotective role of noradrenaline.
Collapse
Affiliation(s)
- Justyna Barut
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Katarzyna Rafa-Zabłocka
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Agnieszka M Jurga
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Monika Bagińska
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Irena Nalepa
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland
| | - Rosanna Parlato
- Division of Neurodegenerative Disorders, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim Heidelberg University, Mannheim, Germany; Institute of Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany.
| | - Grzegorz Kreiner
- Dept. Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, Smętna 12, Poland.
| |
Collapse
|
10
|
Vai B, Mazza MG, Cazzetta S, Calesella F, Aggio V, Lorenzi C, Zanardi R, Poletti S, Colombo C, Benedetti F. Higher Interleukin 13 differentiates patients with a positive history of suicide attempts in major depressive disorder. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2021. [DOI: 10.1016/j.jadr.2021.100254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
11
|
Feng T, Wang J, Cheng K, Lu Q, Zhao R, Wang S, Zhang Q, Ge L, Pan J, Song G, Wang L. IL13Rα1 prevents a castration resistant phenotype of prostate cancer by targeting hexokinase 2 for ubiquitin-mediated degradation. Cancer Biol Med 2021; 19:j.issn.2095-3941.2020.0583. [PMID: 34652890 PMCID: PMC9334759 DOI: 10.20892/j.issn.2095-3941.2020.0583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/02/2021] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Androgen deprivation therapy (ADT) is still the principal treatment option for prostate cancer (PCa). In addition to reactivation of androgen receptor signaling, the resistance of PCa to apoptosis during ADT also contributes to castration resistant PCa (CRPC). A previous study reported that gene transfer of IL-13Rα2 into PCa cells sensitized the cells to the IL-13R-targeted cytotoxin IL13Rα1, leading to apoptosis. Compared with IL-13Rα2, IL13Rα1 is more constitutively expressed in PCa cells, but its function in PCa remains to be established. METHODS We determined the role and expression of IL13Rα1 in PCa cancer cells using western blotting, flow cytometry, and cell proliferation assays. Co-immunoprecipitation and mass spectrometry were used to identify the proteins that interacted with IL13Rα1, to elucidate its function. RESULTS In this study, we showed that IL13Rα1 was selectively suppressed in androgen-deprived PCa cells and that its suppression tended to be associated with poor prognoses of PCa patients. IL13Rα1 overexpression promoted apoptosis and inhibited tumor growth under androgen-deprived or castrated conditions (P < 0.01). Mechanistically, IL13Rα1 recruited and facilitated ubiquitin protein ligase E3C-mediated ubiquitination and degradation of hexokinase 2 (HK2), resulting in glycolytic inhibition and eventually leading to PCa cell apoptosis. Furthermore, our data revealed that mutated ataxia-telangiectasia kinase phosphorylated and facilitated the selective ubiquitin proteasome-mediated degradation of HK2. Notably, IL13Rα1-overexpressing PCa cells were more susceptible to apoptosis and exhibited reduced tumor growth after exposure to the HK2 inhibitor, 2-deoxy-D-glucose (P < 0.01). CONCLUSIONS Our data identified a tumor suppressor role for IL13Rα1 in preventing the resistance of PCa cells to apoptosis during androgen deprivation by inhibiting glycolysis. IL13Rα1-mediated signaling involving HK2 may therefore provide a novel treatment target and strategy for CRPC.
Collapse
Affiliation(s)
- Tingting Feng
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jing Wang
- Department of Pathology, The Fourth People’s Hospital of Jinan, Jinan 250031, China
| | - Kai Cheng
- Department of PET-CT, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250002, China
| | - Qiqi Lu
- The Second Hospital, Cheeloo College of Medicine, Shandong University Medical School, Jinan 250012, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
| | - Ru Zhao
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Shiguan Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
- Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan 250012, China
| | - Qingyun Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
| | - Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250002, China
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| |
Collapse
|
12
|
Yeung SSH, Ho YS, Chang RCC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med 2021; 53:1251-1267. [PMID: 34489558 PMCID: PMC8492689 DOI: 10.1038/s12276-021-00660-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.
Collapse
Affiliation(s)
- Sherry Sin-Hang Yeung
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Yuen-Shan Ho
- grid.16890.360000 0004 1764 6123School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR China
| | - Raymond Chuen-Chung Chang
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| |
Collapse
|
13
|
Bonte MA, El Idrissi F, Gressier B, Devos D, Belarbi K. Protein network exploration prioritizes targets for modulating neuroinflammation in Parkinson's disease. Int Immunopharmacol 2021; 95:107526. [PMID: 33756233 DOI: 10.1016/j.intimp.2021.107526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/20/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is a progressive neurodegenerative disease associated with a loss of dopaminergic neurons in the substantia nigra of the brain. Neuroinflammation, another hallmark of the disease, is thought to play an important role in the neurodegenerative process. While mitigating neuroinflammation could prove beneficial for Parkinson's disease, identifying the most relevant biological processes and pharmacological targets as well as drugs to modulate them remains highly challenging. The present study aimed to better understand the protein network behind neuroinflammation in Parkinson's disease and to prioritize possible targets for its pharmacological modulation. We first used text-mining to systematically collect the proteins significantly associated to Parkinson's disease neuroinflammation over the scientific literature. The functional interaction network formed by these proteins was then analyzed by integrating functional enrichment, network topology analysis and drug-protein interaction analysis. We identified 57 proteins significantly associated to neuroinflammation in Parkinson's disease. Toll-like Receptor Cascades as well as Interleukin 4, Interleukin 10 and Interleukin 13 signaling appeared as the most significantly enriched biological processes. Protein network analysis using STRING and CentiScaPe identified 8 proteins with the highest ability to control these biological processes underlying neuroinflammation, namely caspase 1, heme oxygenase 1, interleukin 1beta, interleukin 4, interleukin 6, interleukin 10, tumor necrosis factor alpha and toll-like receptor 4. These key proteins were indexed to be targetable by a total of 38 drugs including 27 small compounds 11 protein-based therapies. In conclusion, our study highlights key proteins in Parkinson's disease neuroinflammation as well as pharmacological compounds acting on them. As such, it may facilitate the prioritization of biomarkers for the development of diagnostic, target-engagement assessment and therapeutic tools against Parkinson's disease.
Collapse
Affiliation(s)
- Marie-Amandine Bonte
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France.
| | - Fatima El Idrissi
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France.
| | - Bernard Gressier
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France.
| | - David Devos
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie Médicale, I-SITE ULNE, LiCEND, Lille, France.
| | - Karim Belarbi
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France.
| |
Collapse
|
14
|
Picard K, St-Pierre MK, Vecchiarelli HA, Bordeleau M, Tremblay MÈ. Neuroendocrine, neuroinflammatory and pathological outcomes of chronic stress: A story of microglial remodeling. Neurochem Int 2021; 145:104987. [PMID: 33587954 DOI: 10.1016/j.neuint.2021.104987] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
Microglia, the resident macrophage cells of the central nervous system (CNS), are involved in a myriad of processes required to maintain CNS homeostasis. These cells are dynamic and can adapt their phenotype and functions to the physiological needs of the organism. Microglia rapidly respond to changes occurring in their microenvironment, such as the ones taking place during stress. While stress can be beneficial for the organism to adapt to a situation, it can become highly detrimental when it turns chronic. Microglial response to prolonged stress may lead to an alteration of their beneficial physiological functions, becoming either maladaptive or pro-inflammatory. In this review, we aim to summarize the effects of chronic stress exerted on microglia through the neuroendocrine system and inflammation at adulthood. We also discuss how these effects of chronic stress could contribute to microglial involvement in neuropsychiatric and sleep disorders, as well as neurodegenerative diseases.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Marie-Kim St-Pierre
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | | | - Maude Bordeleau
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
15
|
Yang X, Guo Q, Feng T, Lu Q, Ge L, Pan J, Bi K, Qiao L, Tian L, Xie T, Yao C, Song G, Wang L. IL13Rα1 protects against rheumatoid arthritis by combating the apoptotic resistance of fibroblast-like synoviocytes. Arthritis Res Ther 2020; 22:184. [PMID: 32771038 PMCID: PMC7414989 DOI: 10.1186/s13075-020-02270-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 07/13/2020] [Indexed: 12/29/2022] Open
Abstract
Background Endoplasmic reticulum (ER) stress is closely related with the pathological progression of rheumatoid arthritis (RA), and fibroblast-like synoviocytes (FLSs) are known as its resistance against ER stress-induced apoptosis. Studies on overcoming such resistance would provide a novel treatment strategy for RA in a clinical setting. Methods IL13Rα1 expression was assessed in the synovial tissue by RT-qPCR, immunohistology, and Western blot. Gain or loss of functional analysis was applied to evaluate the biological roles of IL13Rα1 in RA FLSs. Cell viability and apoptosis were assessed by MTS, Western blot, and flow cytometry. The therapeutic effects of IL13Rα1 on the severity of type II collagen-induced arthritis (CIA) in DBA-/1 mouse model were evaluated by scoring synovitis, hyperplasia, cartilage degradation, and bone destruction. Results IL13Rα1 expression was selectively downregulated when RA FLSs were stimulated by ER stress inducers. Functionally, IL13Rα1 overexpression could inhibit the viability, but induce the apoptosis of RA FLSs in the presence of ER stress inducers. Mechanistically, IL13Rα1 promotes cell apoptosis via transcriptionally activating trail expression. Besides, IL13Rα1 could interact and stabilize DR5 protein, thus forming a positive loop involving trail and DR5 to render RA FLSs more susceptible to apoptosis. Additionally, intraarticular injection of IL13Rα1 conferred therapeutic effects in CIA models and showed a limited degree of synovial proliferation and joint destruction. Conclusions Together, our data establishes a regulatory role for IL13Rα1 to combat the apoptotic resistance of RA FLSs against ER stress. The inhibitory effects of IL13Rα1 on arthritis progression suggest the therapeutic potential in RA.
Collapse
Affiliation(s)
- Xiaomei Yang
- Department of Hematology, Qilu Children's Hospital of Shandong University, Jinan, China.,Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Qingwei Guo
- Department of Hematology, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Tingting Feng
- Department of Pathology, Shandong University Medical School, Jinan, China
| | - Qiqi Lu
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Jinan, 250062, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China
| | - Luna Ge
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Jinan, 250062, China
| | - Jihong Pan
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Jinan, 250062, China
| | - Kehong Bi
- Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Li Qiao
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lei Tian
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Tianhua Xie
- Department of Rheumatology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Chengfang Yao
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
| | - Lin Wang
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Key Lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong Medicinal Biotechnology Centre, Jinan, 250062, China.
| |
Collapse
|
16
|
Two single nucleotide polymorphisms in IL13 and IL13RA1 from individuals with idiopathic Parkinson's disease increase cellular susceptibility to oxidative stress. Brain Behav Immun 2020; 88:920-924. [PMID: 32276028 PMCID: PMC9012133 DOI: 10.1016/j.bbi.2020.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 01/18/2023] Open
Abstract
The human genes for interleukin 13 (IL-13) and its receptor alpha 1 (IL-13Rα1) are in chromosomal regions associated with Parkinson's disease (PD). The interaction of IL-13 with its receptor increases the susceptibility of mouse dopaminergic neurons to oxidative stress. We identified two rare single SNPs in IL13 and IL13RA1 and measured their cytotoxic effects. rs148077750 is a missense leucine to proline substitution in IL13. It was found in individuals with early onset PD and no other known monogenic forms of the disease and is significantly linked with PD (Fisher's exact test: p-value = 0.01, odds ratio = 14.2). rs145868092 is a leucine to phenylalanine substitution in IL13RA1 affecting a residue critical for IL-13 binding. Both mutations increased the cytotoxic activity of IL-13 on human SH-SY5Y neurons exposed to sublethal doses of hydrogen peroxide, t-butyl hydroperoxide or RLS3, an inducer of ferroptosis. Our data show that both rs148077750 and rs145868092 conferred a gain-of-function that may increase the risk of developing PD.
Collapse
|
17
|
Dong Y, Wang X, Zhou Y, Zheng Q, Chen Z, Zhang H, Sun Z, Xu G, Hu G. Hypothalamus-pituitary-adrenal axis imbalance and inflammation contribute to sex differences in separation- and restraint-induced depression. Horm Behav 2020; 122:104741. [PMID: 32165183 DOI: 10.1016/j.yhbeh.2020.104741] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 02/07/2023]
Abstract
Whether social contact contributes to the underlying mechanisms of depression and the observed sex differences is unclear. In this study, we subjected young male and female mice to separation- and restraint-induced stress for 4 weeks and assessed behaviors, neurotransmitter levels, hormones, and inflammatory cytokines. Results showed that, compared with controls, male mice exposed to stress displayed significant decreases in body weight and sucrose preference after 1 week. In the fourth week, they exhibited a higher degree of anxiety (open field test) and depressive-like behavior (forced swim test). Moreover, the males showed significant decreases in monoamine neurotransmitters, including norepinephrine and dopamine in striatum, and an increase in pro-inflammatory cytokines, such as tumor necrosis factor α and interleukin 1β in serum. In contrast, females showed persistent loss of weight during stress and displayed significant decreases in sucrose preference after stress. Importantly, the females but not males showed activation of the hypothalamus-pituitary-adrenal (HPA) axis, with significantly higher levels adrenocorticotropic hormone. Additionally, mRNA level of c-fos and AVP showed there was significant interaction between stress and sex. Finally, we conclude that an imbalance of the HPA axis and inflammation might be important contributors to sex differences in separation/restraint-induced depressive behavior and that changes might be mediated by c-fos and AVP.
Collapse
Affiliation(s)
- Yinfeng Dong
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xuyang Wang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai 200233, China
| | - Yan Zhou
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiaomu Zheng
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zheng Chen
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hua Zhang
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhiling Sun
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guihua Xu
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
18
|
Nikolenko VN, Gridin LA, Oganesyan MV, Rizaeva NA, Podolskiy YS, Kudryashova VA, Kochurova EV, Kostin RK, Tyagunova EE, Mikhaleva LM, Avila-Rodriguez M, Somasundaram SG, Kirkland CE, Aliev G. The Posterior Perforated Substance: A Brain Mystery Wrapped in an Enigma. Curr Top Med Chem 2020; 19:2991-2998. [PMID: 31775602 DOI: 10.2174/1568026619666191127122452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND There is a dearth of published information on the posterior perforated substance as compared to the anterior perforated substance. We managed to glean facts about the posterior perforated substance that can serve as a landmark for surgical operations in the adjacent regions of the midbrain and the vessels passing through it. Moreover, the posterior perforated substance contains the interpeduncular nucleus responsible for the mental state of the individual. OBJECTIVES 1) To describe the topography of the blood vessels supplying the posterior perforated substance area from the surgical point of view; 2) to investigate the functions of the interpeduncular nucleus. METHODS We assembled and analyzed results from source databases by Elsevier, NCBI MedLine, Scopus, Scholar. Google and Embase. Each article was studied in detail for practically useful information about the posterior perforated substance. RESULTS The P1-segment perforating branches of the posterior cerebral artery supply the posterior perforated substance. This area is especially vulnerable in the case of vascular pathologies. The posterior communicating artery can block the surgeon's view and impede maneuverability of the tool in the area of the posterior perforated substance, which may be addressed using the separation technique, which can lead to positive results. In addition, the medial habenula-interpeduncular nucleus in the posterior perforated substance is associated with various addictions and psychiatric conditions. CONCLUSION The posterior perforated substance area is of great interest for surgical interventions. Future studies of the interpeduncular nucleus anticipate the development of drugs to affect different types of dependencies and some mental diseases.
Collapse
Affiliation(s)
- Vladimir N Nikolenko
- Department of Human Anatomy, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation.,Department of Normal and Topographical Anatomy, Lomonosov Moscow State University, Moscow,Russian Federation
| | - Leonid A Gridin
- Department of Integrative Medicine, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Marine V Oganesyan
- Department of Human Anatomy, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Negoriya A Rizaeva
- Department of Human Anatomy, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Yury S Podolskiy
- Department of Anesthesiology and Resuscitation with the Chambers of Resuscitation and Intensive Therapy No. 2, University Clinical Hospital No. 3, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Valentina A Kudryashova
- Department of Human Anatomy, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Ekaterina V Kochurova
- Department of Prosthetic Dentistry, Dental Institute, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Roman K Kostin
- International School "Medicine of Future" of Biomedical Park of I.M. Sechenov, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Ekaterina E Tyagunova
- International School "Medicine of Future" of Biomedical Park of I.M. Sechenov, First Moscow State Medical University (Sechenov University), Moscow,Russian Federation
| | - Liudmila M Mikhaleva
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418,Russian Federation
| | - Marco Avila-Rodriguez
- Department of Clinic Sciences, Faculty of Medicine. University of Tolima, Ibagué - 730001,Colombia
| | - Siva G Somasundaram
- Department of Biological Sciences, Salem University, 223 West Main Street Salem, WV 26426,United States
| | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, 223 West Main Street Salem, WV 26426,United States
| | - Gjumrakch Aliev
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418,Russian Federation.,I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya str., Moscow, 119991,Russian Federation.,Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432,Russian Federation.,GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229,United States
| |
Collapse
|
19
|
Miao W, Zhao Y, Huang Y, Chen D, Luo C, Su W, Gao Y. IL-13 Ameliorates Neuroinflammation and Promotes Functional Recovery after Traumatic Brain Injury. THE JOURNAL OF IMMUNOLOGY 2020; 204:1486-1498. [PMID: 32034062 DOI: 10.4049/jimmunol.1900909] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/05/2020] [Indexed: 12/16/2022]
Abstract
Microglia play essential roles in neuroinflammatory responses after traumatic brain injury (TBI). Our previous studies showed that phenotypes of microglia, as well as infiltrating macrophages, altered at different stages after CNS injury, which was correlated to functional outcomes. IL-13 is an anti-inflammatory cytokine that has been reported to protect against demyelination and spinal cord injury through immunomodulation. The effects of IL-13 in microglia/macrophage-mediated immune responses after TBI remain unknown. In this study, we showed that intranasal administration of IL-13 in male C57BL/6J mice accelerated functional recovery in the controlled cortical impact model of TBI. IL-13 treatment increased the time to fall off in the Rotarod test, reduced the number of foot faults in the foot fault test, and improved the score in the wire hang test up to 28 d after TBI. Consistent with functional improvement, IL-13 reduced neuronal tissue loss and preserved white matter integrity 6 d after TBI. Furthermore, IL-13 ameliorated the elevation of proinflammatory factors and reduced the number of proinflammatory microglia/macrophages 6 d after TBI. Additionally, IL-13 enhanced microglia/macrophage phagocytosis of damaged neurons in the peri-lesion areas. In vitro studies confirmed that IL-13 treatment inhibited the production of proinflammatory cytokines in rat primary microglia in response to LPS or dead neuron stimulation and increased the ability of microglia to engulf fluorophore-labeled latex beads or dead neurons. Collectively, we demonstrated that IL-13 treatment improved neurologic outcomes after TBI through adjusting microglia/macrophage phenotypes and inhibiting inflammatory responses. IL-13 may represent a potential immunotherapy to promote long-term recovery from TBI.
Collapse
Affiliation(s)
- Wanying Miao
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Yongfang Zhao
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Di Chen
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Chen Luo
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Wei Su
- Department of Neurosurgery, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; .,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| |
Collapse
|
20
|
Sanchez-Alavez M, Nguyen W, Mori S, Wills DN, Otero D, Aguirre CA, Singh M, Ehlers CL, Conti B. Time Course of Blood and Brain Cytokine/Chemokine Levels Following Adolescent Alcohol Exposure and Withdrawal in Rats. Alcohol Clin Exp Res 2019; 43:2547-2558. [PMID: 31589333 PMCID: PMC6904424 DOI: 10.1111/acer.14209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 10/01/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Adolescence is a critical period for neural development, and alcohol exposure during adolescence can lead to an elevated risk for health consequences as well as alcohol use disorders. Clinical and experimental data suggest that chronic alcohol exposure may produce immunomodulatory effects that can lead to the activation of pro-inflammatory cytokine pathways as well as microglial markers. The present study evaluated, in brain and blood, the effects of adolescent alcohol exposure and withdrawal on microglia and on the most representative pro- and anti-inflammatory cytokines and major chemokines that can contribute to the establishing of a neuroinflammatory environment. METHODS Wistar rats (males, n = 96) were exposed to ethanol (EtOH) vapors, or air control, for 5 weeks over adolescence (PD22-PD58). Brains and blood samples were collected at 3 time points: (i) after 35 days of vapor/air exposure (PD58); (ii) after 1 day of withdrawal (PD59), and (iii) 28 days after withdrawal (PD86). The ionized calcium-binding adapter molecule 1 (Iba-1) was used to index microglial activation, and cytokine/chemokine responses were analyzed using magnetic bead panels. RESULTS After 35 days of adolescent vapor exposure, a significant increase in Iba-1 immunoreactivity was seen in amygdala, frontal cortex, hippocampus, and substantia nigra. However, Iba-1 density returned to control levels at both 1 day and 28 days of withdrawal except in the hippocampus where Iba-1 density was significantly lower than controls. In serum, adolescent EtOH exposure induced a reduction in IL-13 and an increase in fractalkine at day 35. After 1 day of withdrawal, IL-18 was reduced, and IP-10 was elevated, whereas both IP-10 and IL-10 were elevated at 28 days following withdrawal. In the frontal cortex, adolescent EtOH exposure induced an increase in IL-1β at day 35, and 28 days of withdrawal, and IL-10 was increased after 28 days of withdrawal. CONCLUSION These data demonstrate that EtOH exposure during adolescence produces significant microglial activation; however, inflammatory markers seen in the blood appear to differ from those observed in the brain.
Collapse
Affiliation(s)
| | - William Nguyen
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Simone Mori
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Derek N Wills
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Dennis Otero
- Infectious and Inflammatory Disease Center and National Cancer Institute (NCI)-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Research Institute, La Jolla, California
| | - Carlos A Aguirre
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Mona Singh
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Cindy L Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Bruno Conti
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
21
|
Furuyashiki T, Kitaoka S. Neural mechanisms underlying adaptive and maladaptive consequences of stress: Roles of dopaminergic and inflammatory responses. Psychiatry Clin Neurosci 2019; 73:669-675. [PMID: 31215710 DOI: 10.1111/pcn.12901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/03/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022]
Abstract
Stress caused by adverse and demanding conditions, a risk factor for mental illnesses, induces adaptive or maladaptive neural and behavioral consequences, depending on the conditions. Studies using rodent stress models have revealed multiple mechanisms related to dopamine and inflammation for stress-induced neural and behavioral changes. Thus, repeated stress alters activities of ventral tegmental area dopamine neurons projecting to the nucleus accumbens and the medial prefrontal cortex in distinct manners. In the nucleus accumbens, repeated stress decreases activities of D1 receptor-expressing neurons. In the medial prefrontal cortex, single stress increases dopamine D1 receptor signaling, leading to dendritic hypertrophy of excitatory neurons and stress resilience. These changes are attenuated with repetition of stress via prostaglandin E2 , an inflammation-related lipid mediator. Repeated stress activates microglia in the medial prefrontal cortex and the hippocampus. Innate immune receptors, such as the toll-like receptor 2/4 and P2X7, are crucial for repeated stress-induced microglial activation, leading to neural and behavioral changes through proinflammatory cytokines. In addition, repeated stress induces monocyte infiltration to the brain, and impairs the blood-brain barrier in the nucleus accumbens, leading to cytokine leakage to the brain. These monocyte-derived responses are involved in stress-induced behavioral changes. These findings show crucial roles of the accumbal and prefrontal dopamine pathways and inflammatory responses in the brain and body to direct adaptive and maladaptive consequences of stress, and pave the way for identifying a neural origin of stress and understanding the stress-related pathology of mental illnesses.
Collapse
Affiliation(s)
- Tomoyuki Furuyashiki
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Japan.,Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Shiho Kitaoka
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Japan.,Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
22
|
Olde Heuvel F, Holl S, Chandrasekar A, Li Z, Wang Y, Rehman R, Förstner P, Sinske D, Palmer A, Wiesner D, Ludolph A, Huber-Lang M, Relja B, Wirth T, Röszer T, Baumann B, Boeckers T, Knöll B, Roselli F. STAT6 mediates the effect of ethanol on neuroinflammatory response in TBI. Brain Behav Immun 2019; 81:228-246. [PMID: 31207335 DOI: 10.1016/j.bbi.2019.06.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/31/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) and ethanol intoxication (EI) frequently coincide, particularly in young subjects. However, the mechanisms of their interaction remain poorly understood. Among other pathogenic pathways, TBI induces glial activation and neuroinflammation in the hippocampus, resulting in acute and chronic hippocampal dysfunction. In this regard, we investigated the role of EI affecting these responses unfolding after TBI. We used a blunt, weight-drop approach to model TBI in mice. Male mice were pre-administered with ethanol or vehicle to simulate EI. The neuroinflammatory response in the hippocampus was assessed by monitoring the expression levels of >20 cytokines, the phosphorylation status of transcription factors and the phenotype of microglia and astrocytes. We used AS1517499, a brain-permeable STAT6 inhibitor, to elucidate the role of this pathway in the EI/TBI interaction. We showed that TBI causes the elevation of IL-33, IL-1β, IL-38, TNF-α, IFN-α, IL-19 in the hippocampus at 3 h time point and concomitant EI results in the dose-dependent downregulation of IL-33, IL-1β, IL-38, TNF-α and IL-19 (but not of IFN-α) and in the selective upregulation of IL-13 and IL-12. EI is associated with the phosphorylation of STAT6 and the transcription of STAT6-controlled genes. Moreover, ethanol-induced STAT6 phosphorylation and transcriptional activation can be recapitulated in vitro by concomitant exposure of neurons to ethanol, depolarization and inflammatory stimuli (simulating the acute trauma). Acute STAT6 inhibition prevents the effects of EI on IL-33 and TNF-α, but not on IL-13 and negates acute EI beneficial effects on TBI-associated neurological impairment. Additionally, EI is associated with reduced microglial activation and astrogliosis as well as preserved synaptic density and baseline neuronal activity 7 days after TBI and all these effects are prevented by acute administration of the STAT6 inhibitor concomitant to EI. EI concomitant to TBI exerts significant immunomodulatory effects on cytokine induction and microglial activation, largely through the activation of STAT6 pathway, ultimately with beneficial outcomes.
Collapse
Affiliation(s)
- Florian Olde Heuvel
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Sarah Holl
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Akila Chandrasekar
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Zhenghui Li
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Yibin Wang
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Rida Rehman
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Philip Förstner
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Daniela Sinske
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Diana Wiesner
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Albert Ludolph
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Borna Relja
- Dept. of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Tamás Röszer
- Institute of Neurobiology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Tobias Boeckers
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany; Institute of Anatomy and Cell Biology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Francesco Roselli
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany; Institute of Anatomy and Cell Biology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
23
|
Ethanol Induction of Innate Immune Signals Across BV2 Microglia and SH-SY5Y Neuroblastoma Involves Induction of IL-4 and IL-13. Brain Sci 2019; 9:brainsci9090228. [PMID: 31510019 PMCID: PMC6770440 DOI: 10.3390/brainsci9090228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023] Open
Abstract
Innate immune signaling molecules, such as Toll-like receptors (TLRs), cytokines and transcription factor NFκB, are increased in post-mortem human alcoholic brain and may play roles in alcohol dependence and neurodegeneration. Innate immune signaling involves microglia -neuronal signaling which while poorly understood, may impact learning and memory. To investigate mechanisms of ethanol induction of innate immune signaling within and between brain cells, we studied immortalized BV2 microglia and SH-SY5Y human neuroblastoma to model microglial and neuronal signaling. Cells were treated alone or in co-culture using a Transwell system, which allows transfer of soluble mediators. We determined immune signaling mRNA using real-time polymerase chain reaction. Ethanol induced innate immune genes in both BV2 and SH-SY5Y cultured alone, with co-culture altering gene expression at baseline and following ethanol exposure. Co-culture blunted ethanol-induced high mobility group box protein 1 (HMGB1)-TLR responses, corresponding with reduced ethanol induction of several proinflammatory NFκB target genes. In contrast, co-culture resulted in ethanol upregulation of cytokines IL-4 and IL-13 in BV2 and corresponding receptors, that is, IL-4 and IL-13 receptors, in SH-SY5Y, suggesting induction of a novel signaling pathway. Co-culture reduction in HMGB1-TLR levels occurs in parallel with reduced proinflammatory gene induction and increased IL-4 and IL-13 ligands and receptors. Findings from these immortalized and tumor-derived cell lines could provide insight into microglial-neuronal interactions via release of soluble mediators in vivo.
Collapse
|
24
|
Bok E, Cho EJ, Chung ES, Shin WH, Jin BK. Interleukin-4 Contributes to Degeneration of Dopamine Neurons in the Lipopolysaccharide-treated Substantia Nigra in vivo. Exp Neurobiol 2018; 27:309-319. [PMID: 30181693 PMCID: PMC6120964 DOI: 10.5607/en.2018.27.4.309] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 01/08/2023] Open
Abstract
The present study investigated the effects of interleukin (IL)-4 on dopamine (DA) neurons in the substantia nigra (SN) in vivo of lipopolysaccharide (LPS)-treated rat. Tyrosine hydroxylase immunohistochemistry showed a significant loss of nigral DA neurons at 3 and 7 day post-LPS. In parallel, IL-4 immunoreactivity was upregulated as early as 1 day, reached a peak at 3 day and remained elevated at 7 day post-LPS. IL-4 immunoreactivity was detected exclusively in microglia. IL-4 neutralizing antibody (NA) significantly increased survival of DA neurons in LPS-treated SN in vivo by inhibiting microglial activation and production of proinflammatory mediator such as IL-1β as assessed by immunihistochemical, RT-PCR and ELISA analysis, respectively. Accompanying neuroprotection are IL-4NA effects on decreased disruption of blood-brain barrier and astrocytes. The present data suggest that endogenously expressed IL-4 from reactive microglia may be involved in the neuropathological processes of degeneration of DA neurons occurring in Parkinson's disease.
Collapse
Affiliation(s)
- Eugene Bok
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Eun Ju Cho
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Eun Sook Chung
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Won-Ho Shin
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Byung Kwan Jin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
25
|
Maher P, Conti B. Deciphering the pathways that protect from IL-13-mediated potentiation of oxidative stress-induced dopaminergic nerve cell death. Cytokine 2018; 103:114-120. [PMID: 28969943 PMCID: PMC5808859 DOI: 10.1016/j.cyto.2017.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/31/2017] [Accepted: 09/18/2017] [Indexed: 01/27/2023]
Abstract
The majority of Parkinson's disease (PD) cases are sporadic with only about 10% of PD patients having a family history of the disease suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Both oxidative stress and neuroinflammation are thought to contribute to PD. Previously, we showed that the activation of interleukin 13 receptor alpha 1 (IL-13Rα1) increases the sensitivity of dopaminergic neurons to oxidative damage both in cultured cells and in animals. In this study, we investigated the pathways involved in the IL-13-mediated potentiation of oxidative stress-induced dopaminergic cell death using a combination of cell survival assays and Western blotting with appropriate antibodies. In addition, siRNA was used to examine the role of 4E-BP1 in this cell toxicity paradigm. We show that activation of both the Jak-Stat and PI3 kinase-mTOR pathways play key roles in the promotion of cell death by IL-13 in the presence of mild oxidative stress. The Jak 1/2 inhibitor ruxolitinib, the mTOR inhibitor rapamycin and the PI3 kinase inhibitor LY294002 all prevented the potentiation of cell death by IL-13. Moreover, 4E-BP1, a target of mTOR, appeared to mediate the protective effects of rapamycin. Together, these results indicate that multiple signaling pathways downstream of IL-13Rα1 activation play a role in the toxic effects of IL-13 in dopaminergic neurons in the presence of mild oxidative stress and suggest that any of these pathways might provide potential targets for the treatment of PD.
Collapse
Affiliation(s)
- Pamela Maher
- The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, United States.
| | - Bruno Conti
- The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, United States
| |
Collapse
|