1
|
Rinaldi A, Balietti M, Principi E, De Luca M, De Felice E, Narcisi FM, Vilardo L, Rosito M, Piacentini R, D'Alessandro G, D'Agnano I, Maggi L, Conti F, Limatola C, Catalano M. BV2-derived extracellular vesicles modulate microglia inflammatory profile, neuronal plasticity, and behavioural performances in late adult mice. Brain Behav Immun 2024; 122:58-74. [PMID: 39128568 DOI: 10.1016/j.bbi.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND During aging, both the brain and the immune system undergo a progressive impairment of physiological functions. Microglia, the immunocompetent cells of the central nervous system, shift towards a chronic mild inflammatory state that impacts brain homeostasis. Extracellular vesicles (EVs) released by microglia transport packages of molecular information that mirror the inflammatory status of donor cells and modulate the inflammatory phenotype of recipient microglia and other cell types. RESULTS We demonstrated that intranasal administration of EVs derived from microglial-like BV2 cells to late adult mice (16-20 months of age) shifts microglia toward a "juvenile" morphology affecting their inflammatory profile. Mice treated with BV2-derived EVs have a reduction of anxiety-like behavior and an increased spatial learning, with sex-dependent differences. Further, BV2-derived EVs increased neuronal plasticity both in male and female mice. These findings suggest the involvement of microglial cells in vesicles-mediated anti-aging effect. CONCLUSIONS Our data indicate that BV2-derived EVs could represent a resource to slow down age-dependent inflammation in the mouse brain.
Collapse
Affiliation(s)
- Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Marta Balietti
- IRCCS INRCA, Center for Neurobiology of Aging, Via Birarelli 8, Ancona 60121, Italy
| | - Elisa Principi
- Università Politecnica delle Marche, Department of Experimental and Clinical Medicine, Via Tronto 10/a, Ancona 60126, Italy
| | | | - Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Laura Vilardo
- Institute of Biomedical Technologies, CNR, 20054 Segrate, Italy
| | - Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; Center for Life Nanoscience & Neuroscience Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli 1, Roma, Italy
| | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Igea D'Agnano
- Institute of Biomedical Technologies, CNR, 20054 Segrate, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Fiorenzo Conti
- Università Politecnica delle Marche, Department of Experimental and Clinical Medicine, Via Tronto 10/a, Ancona 60126, Italy; IRCCS INRCA, Center for Neurobiology of Aging, Via Birarelli 8, Ancona 60121, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia, Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
| |
Collapse
|
2
|
Rymbai E, Sugumar D, Chakkittukandiyil A, Kothandan R, Selvaraj D. Molecular insights into the potential effects of selective estrogen receptor β agonists in Alzheimer's and Parkinson's diseases. Cell Biochem Funct 2024; 42:e4014. [PMID: 38616346 DOI: 10.1002/cbf.4014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative disorders. Pathologically, AD and PD are characterized by the accumulation of misfolded proteins. Hence, they are also called as proteinopathy diseases. Gender is considered as one of the risk factors in both diseases. Estrogens are widely accepted to be neuroprotective in several neurodegenerative disorders. Estrogens can be produced in the central nervous system, where they are called as neurosteroids. Estrogens mediate their neuroprotective action mainly through their actions on estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). However, ERα is mainly involved in the growth and development of the primary and secondary sexual organs in females. Hence, the activation of ERα is associated with undesired side effects such as gynecomastia and increase in the risk of breast cancer, thromboembolism, and feminization. Therefore, selective activation of ERβ is often considered to be safer. In this review, we explore the role of ERβ in regulating the expression and functions of AD- and PD-associated genes. Additionally, we discuss the association of these genes with the amyloid-beta peptide (Aβ) and α-synuclein mediated toxicity. Ultimately, we established a correlation between the importance of ERβ activation and the process underlying ERβ's neuroprotective mechanisms in AD and PD.
Collapse
Affiliation(s)
- Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Ram Kothandan
- Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| |
Collapse
|
3
|
Hong Y, Jiang L, Tang F, Zhang M, Cui L, Zhong H, Xu F, Li M, Chen C, Chen L. PPAR-γ promotes the polarization of rat retinal microglia to M2 phenotype by regulating the expression of CD200-CD200R1 under hypoxia. Mol Biol Rep 2023; 50:10277-10285. [PMID: 37971567 DOI: 10.1007/s11033-023-08815-5] [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: 06/10/2023] [Accepted: 09/11/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Recent reports suggest that peroxisome proliferator-activated receptor-γ (PPAR-γ) could promote microglial M2 polarization to inhibit inflammation. However, the specific molecular mechanisms that trigger PPAR-γ's anti-inflammatory ability in microglia are yet to be expounded. Thus, in this study, we aimed to explore the molecular mechanisms behind the anti-inflammatory effects of PPAR-γ in hypoxia-stimulated rat retinal microglial cells. METHODS AND RESULTS We used shRNA expressing lentivirus to knock down PPAR-γ and CD200 genes, and we assessed hypoxia-induced polarization markers release - M1 (iNOS, IL-1β, IL-6, and TNF-α) and M2 (Arg-1, YM1, IL-4, and IL-10) by RT-PCR. We also monitored PPAR-γ-related signals (PPAR-γ, PPAR-γ in cytoplasm or nucleus, CD200, and CD200Rs) by Western blot and RT-PCR. Our results showed that hypoxia enhanced PPAR-γ and CD200 expressions in microglial cells. Moreover, PPAR-γ agonist 15d-PGJ2 elevated CD200 and CD200R1 expressions, whereas sh-PPAR-γ had the opposite effect. Following hypoxia, expressions of M1 markers increased significantly, while those of M2 markers decreased, and the above effects were attenuated by 15d-PGJ2. Conversely, knocking down PPAR-γ or CD200 inhibited the polarization of microglial cells to M2 phenotype. CONCLUSION Our findings demonstrated that PPAR-γ performed an anti-inflammatory function in hypoxia-stimulated microglial cells by promoting their polarization to M2 phenotype via the CD200-CD200R1 pathway.
Collapse
Affiliation(s)
- Yiyi Hong
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Jiang
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Fen Tang
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Mingyuan Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Ling Cui
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Haibin Zhong
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Fan Xu
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Min Li
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Changzheng Chen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Lifei Chen
- Research center of Ophthalmology, Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology & Department of Ophthalmology, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| |
Collapse
|
4
|
Marciante AB, Mitchell GS. Increased spinal adenosine impairs phrenic long-term facilitation in aging rats. J Appl Physiol (1985) 2023; 134:1537-1548. [PMID: 37167263 PMCID: PMC10281789 DOI: 10.1152/japplphysiol.00197.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023] Open
Abstract
Moderate acute intermittent hypoxia (mAIH) elicits a form of spinal, respiratory motor plasticity known as phrenic long-term facilitation (pLTF). In middle-aged male and geriatric female rats, mAIH-induced pLTF is attenuated through unknown mechanisms. In young adults, mAIH activates competing intracellular signaling cascades, initiated by serotonin 2 and adenosine 2A (A2A) receptors, respectively. Spinal A2A receptor inhibition enhances mAIH-induced pLTF, meaning, serotonin dominates, and adenosine constrains mAIH-induced plasticity in the daily rest phase. Thus, we hypothesized elevated basal adenosine levels in the ventral cervical spinal cord of aged rats shifts this balance, undermining mAIH-induced pLTF. A selective A2A receptor antagonist (MSX-3) or vehicle was delivered intrathecally at C4 in anesthetized young (3-6 mo) and aged (20-22 mo) Sprague-Dawley rats before mAIH (3,5-min episodes; arterial Po2 = 45-55 mmHg). In young males, spinal A2A receptor inhibition enhanced pLTF (119 ± 5%) vs. vehicle (55 ± 9%), consistent with prior reports. In old males, pLTF was reduced to 25 ± 11%, but A2A receptor inhibition increased pLTF to levels greater than in young males (186 ± 19%). Basal adenosine levels in ventral C3-C5 homogenates are elevated two- to threefold in old vs. young males. These findings advance our understanding of age as a biological variable in phrenic motor plasticity and will help guide translation of mAIH as a therapeutic modality to restore respiratory and nonrespiratory movements in older populations afflicted with clinical disorders that compromise movement.NEW & NOTEWORTHY Advanced age undermines respiratory motor plasticity, specifically phrenic long-term facilitation (pLTF) following moderate acute intermittent hypoxia (mAIH). We report that spinal adenosine increases in aged male rats, undermining mAIH-induced pLTF via adenosine 2A (A2A) receptor activation, an effect reversed by selective spinal adenosine 2A receptor inhibition. These findings advance our understanding of mechanisms that impair neuroplasticity, and the ability to compensate for the onset of lung or neural injury with age, and may guide efforts to harness mAIH as a treatment for clinical disorders that compromise breathing and other movements.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Department of Physical Therapy & McKnight Brain Institute, Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
| | - Gordon S Mitchell
- Department of Physical Therapy & McKnight Brain Institute, Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
5
|
Abbasloo E, Amiresmaili S, Shirazpour S, Khaksari M, Kobeissy F, Thomas TC. Satureja khuzistanica Jamzad essential oil and pure carvacrol attenuate TBI-induced inflammation and apoptosis via NF-κB and caspase-3 regulation in the male rat brain. Sci Rep 2023; 13:4780. [PMID: 36959464 PMCID: PMC10036533 DOI: 10.1038/s41598-023-31891-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 03/20/2023] [Indexed: 03/25/2023] Open
Abstract
Traumatic brain injury (TBI) causes progressive dysfunction that induces biochemical and metabolic changes that lead to cell death. Nevertheless, there is no definitive FDA-approved therapy for TBI treatment. Our previous immunohistochemical results indicated that the cost-effective natural Iranian medicine, Satureja khuzistanica Jamzad essential oil (SKEO), which consists of 94.16% carvacrol (CAR), has beneficial effects such as reducing neuronal death and inflammatory markers, as well as activating astrocytes and improving neurological outcomes. However, the molecular mechanisms of these neuroprotective effects have not yet been elucidated. This study investigated the possible mechanisms involved in the anti-inflammatory and anti-apoptotic properties of SKEO and CAR after TBI induction. Eighty-four male Wistar rats were randomly divided into six groups: Sham, TBI, TBI + Vehicle, TBI + CAR (100 and 200 mg/kg), and TBI + SKEO (200 mg/kg) groups. After establishing the "Marmarou" weight drop model, diffuse TBI was induced in the rat brain. Thirty minutes after TBI induction, SKEO & CAR were intraperitoneally injected. One day after TBI, injured rats exhibited significant brain edema, neurobehavioral dysfunctions, and neuronal apoptosis. Western blot results revealed upregulation of the levels of cleaved caspase-3, NFκB p65, and Bax/Bcl-2 ratio, which was attenuated by CAR and SKEO (200 mg/kg). Furthermore, the ELISA results showed that CAR treatment markedly prevents the overproduction of the brain pro-inflammatory cytokines, including IL-1β, TNF-α, and IL-6. Moreover, the neuron-specific enolase (NSE) immunohistochemistry results revealed the protective effect of CAR and SKEO on post-TBI neuronal death. The current study revealed that the possible neuroprotective mechanisms of SKEO and CAR might be related to (at least in part) modulating NF-κB regulated inflammation and caspase-3 protein expression. It also suggested that CAR exerts more potent protective effects than SKEO against TBI. Nevertheless, the administration of SKEO and CAR may express a novel therapeutic approach to ameliorate TBI-related secondary phase neuropathological outcomes.
Collapse
Affiliation(s)
- Elham Abbasloo
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | | | - Sara Shirazpour
- Department of Physiology and Pharmacology, Faculty of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Neurotrauma, Multiomics and Biomarkers, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, USA
- Translational Neurotrauma Research Program, Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, USA
| |
Collapse
|
6
|
Arik E, Heinisch O, Bienert M, Gubeljak L, Slowik A, Reich A, Schulz JB, Wilhelm T, Huber M, Habib P. Erythropoietin Enhances Post-ischemic Migration and Phagocytosis and Alleviates the Activation of Inflammasomes in Human Microglial Cells. Front Cell Neurosci 2022; 16:915348. [PMID: 35813499 PMCID: PMC9263298 DOI: 10.3389/fncel.2022.915348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022] Open
Abstract
Recombinant human erythropoietin (rhEPO) has been shown to exert anti-apoptotic and anti-inflammatory effects after cerebral ischemia. Inflammatory cytokines interleukin-1β and -18 (IL-1β and IL-18) are crucial mediators of apoptosis and are maturated by multiprotein complexes termed inflammasomes. Microglia are the first responders to post-ischemic brain damage and are a main source of inflammasomes. However, the impact of rhEPO on microglial activation and the subsequent induction of inflammasomes after ischemia remains elusive. To address this, we subjected human microglial clone 3 (HMC-3) cells to various durations of oxygen-glucose-deprivation/reperfusion (OGD/R) to assess the impact of rhEPO on cell viability, metabolic activity, oxidative stress, phagocytosis, migration, as well as on the regulation and activation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes. Administration of rhEPO mitigated OGD/R-induced oxidative stress and cell death. Additionally, it enhanced metabolic activity, migration and phagocytosis of HMC-3. Moreover, rhEPO attenuated post-ischemic activation and regulation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes as well as their downstream effectors CASPASE1 and IL-1β. Pharmacological inhibition of NLRP3 via MCC950 had no effect on the activation of CASPASE1 and maturation of IL-1β after OGD/R, but increased protein levels of NLRP1, NLRC4, and AIM2, suggesting compensatory activities among inflammasomes. We provide evidence that EPO-conveyed anti-inflammatory actions might be mediated via the regulation of the inflammasomes.
Collapse
Affiliation(s)
- Eren Arik
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ole Heinisch
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michaela Bienert
- Institute of Molecular and Cellular Anatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Lara Gubeljak
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jörg B. Schulz
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
- *Correspondence: Pardes Habib, ; orcid.org/0000-0002-5771-216X
| |
Collapse
|
7
|
Mitchell GS, Baker TL. Respiratory neuroplasticity: Mechanisms and translational implications of phrenic motor plasticity. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:409-432. [PMID: 35965036 DOI: 10.1016/b978-0-323-91534-2.00016-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Widespread appreciation that neuroplasticity is an essential feature of the neural system controlling breathing has emerged only in recent years. In this chapter, we focus on respiratory motor plasticity, with emphasis on the phrenic motor system. First, we define related but distinct concepts: neuromodulation and neuroplasticity. We then focus on mechanisms underlying two well-studied models of phrenic motor plasticity: (1) phrenic long-term facilitation following brief exposure to acute intermittent hypoxia; and (2) phrenic motor facilitation after prolonged or recurrent bouts of diminished respiratory neural activity. Advances in our understanding of these novel and important forms of plasticity have been rapid and have already inspired translation in multiple respects: (1) development of novel therapeutic strategies to preserve/restore breathing function in humans with severe neurological disorders, such as spinal cord injury and amyotrophic lateral sclerosis; and (2) the discovery that similar plasticity also occurs in nonrespiratory motor systems. Indeed, the realization that similar plasticity occurs in respiratory and nonrespiratory motor neurons inspired clinical trials to restore leg/walking and hand/arm function in people living with chronic, incomplete spinal cord injury. Similar application may be possible to other clinical disorders that compromise respiratory and non-respiratory movements.
Collapse
Affiliation(s)
- Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| | - Tracy L Baker
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
8
|
Guo L, Zhu L. Multiple Roles of Peripheral Immune System in Modulating Ischemia/Hypoxia-Induced Neuroinflammation. Front Mol Biosci 2021; 8:752465. [PMID: 34881289 PMCID: PMC8645603 DOI: 10.3389/fmolb.2021.752465] [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: 08/03/2021] [Accepted: 10/05/2021] [Indexed: 12/20/2022] Open
Abstract
Given combined efforts of neuroscience and immunology, increasing evidence has revealed the critical roles of the immune system in regulating homeostasis and disorders of the central nervous system (CNS). Microglia have long been considered as the only immune cell type in parenchyma, while at the interface between CNS and the peripheral (meninges, choroid plexus, and perivascular space), embryonically originated border-associated macrophages (BAMs) and multiple surveilling leukocytes capable of migrating into and out of the brain have been identified to function in the healthy brain. Hypoxia-induced neuroinflammation is the key pathological procedure that can be detected in healthy people at high altitude or in various neurodegenerative diseases, during which a very thin line between a beneficial response of the peripheral immune system in maintaining brain homeostasis and a pathological role in exacerbating neuroinflammation has been revealed. Here, we are going to focus on the role of the peripheral immune system and its crosstalk with CNS in the healthy brain and especially in hypobaric or ischemic hypoxia-associated neuroinflammation.
Collapse
Affiliation(s)
- Liang Guo
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lingling Zhu
- Beijing Institute of Basic Medical Sciences, Beijing, China.,University of Nanhua, Hengyang, China.,Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Nuclear Receptors in Myocardial and Cerebral Ischemia-Mechanisms of Action and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms222212326. [PMID: 34830207 PMCID: PMC8617737 DOI: 10.3390/ijms222212326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.
Collapse
|
10
|
Erythropoietin Abrogates Post-Ischemic Activation of the NLRP3, NLRC4, and AIM2 Inflammasomes in Microglia/Macrophages in a TAK1-Dependent Manner. Transl Stroke Res 2021; 13:462-482. [PMID: 34628598 PMCID: PMC9046144 DOI: 10.1007/s12975-021-00948-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/18/2021] [Accepted: 09/18/2021] [Indexed: 12/18/2022]
Abstract
Inflammasomes are known to contribute to brain damage after acute ischemic stroke (AIS). TAK1 is predominantly expressed in microglial cells and can regulate the NLRP3 inflammasome, but its impact on other inflammasomes including NLRC4 and AIM2 after AIS remains elusive. EPO has been shown to reduce NLRP3 protein levels in different disease models. Whether EPO-mediated neuroprotection after AIS is conveyed via an EPO/TAK1/inflammasome axis in microglia remains to be clarified. Subjecting mice deficient for TAK1 in microglia/macrophages (Mi/MΦ) to AIS revealed a significant reduction in infarct sizes and neurological impairments compared to the corresponding controls. Post-ischemic increased activation of TAK1, NLRP3, NLRC4, and AIM2 inflammasomes including their associated downstream cascades were markedly reduced upon deletion of Mi/MΦ TAK1. EPO administration improved clinical outcomes and dampened stroke-induced activation of TAK1 and inflammasome cascades, which was not evident after the deletion of Mi/MΦ TAK1. Pharmacological inhibition of NLRP3 in microglial BV-2 cells did not influence post-OGD IL-1β levels, but increased NLRC4 and AIM2 protein levels, suggesting compensatory activities among inflammasomes. Overall, we provide evidence that Mi/MΦ TAK1 regulates the expression and activation of the NLRP3, NLRC4, AIM2 inflammasomes. Furthermore, EPO mitigated stroke-induced activation of TAK1 and inflammasomes, indicating that EPO conveyed neuroprotection might be mediated via an EPO/TAK1/inflammasome axis.
Collapse
|
11
|
Xu J, Zhou Y, Yan C, Wang X, Lou J, Luo Y, Gao S, Wang J, Wu L, Gao X, Shao A. Neurosteroids: A novel promise for the treatment of stroke and post-stroke complications. J Neurochem 2021; 160:113-127. [PMID: 34482541 DOI: 10.1111/jnc.15503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023]
Abstract
Stroke is the primary reason for death and disability worldwide, with few treatment strategies to date. Neurosteroids, which are natural molecules in the brain, have aroused great interest in the field of stroke. Neurosteroids are a kind of steroid that acts on the nervous system, and are synthesized in the mitochondria of neurons or glial cells using cholesterol or other steroidal precursors. Neurosteroids mainly include estrogen, progesterone (PROG), allopregnanolone, dehydroepiandrosterone (DHEA), and vitamin D (VD). Most of the preclinical studies have confirmed that neurosteroids can decrease the risk of stroke, and improve stroke outcomes. In the meantime, neurosteroids have been shown to have a positive therapeutic significance in some post-stroke complications, such as epilepsy, depression, anxiety, cardiac complications, movement disorders, and post-stroke pain. In this review, we report the historical background, modulatory mechanisms of neurosteroids in stroke and post-stroke complications, and emphasize on the application prospect of neurosteroids in stroke therapy.
Collapse
Affiliation(s)
- Jiawei Xu
- The First Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Caochong Yan
- The Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianyao Lou
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Luo
- The Second Affiliated Hospital of Zhejiang University School of Medicine (Changxing Branch), Changxing, Huzhou, Zhejiang, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junjie Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liang Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangfu Gao
- The First Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Connor M, Lamorie-Foote K, Liu Q, Shkirkova K, Baertsch H, Sioutas C, Morgan TE, Finch CE, Mack WJ. Nanoparticulate matter exposure results in white matter damage and an inflammatory microglial response in an experimental murine model. PLoS One 2021; 16:e0253766. [PMID: 34214084 PMCID: PMC8253444 DOI: 10.1371/journal.pone.0253766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/14/2021] [Indexed: 01/25/2023] Open
Abstract
Exposure to ambient air pollution has been associated with white matter damage and neurocognitive decline. However, the mechanisms of this injury are not well understood and remain largely uncharacterized in experimental models. Prior studies have shown that exposure to particulate matter (PM), a sub-fraction of air pollution, results in neuroinflammation, specifically the upregulation of inflammatory microglia. This study examines white matter and axonal injury, and characterizes microglial reactivity in the corpus callosum of mice exposed to 10 weeks (150 hours) of PM. Nanoscale particulate matter (nPM, aerodynamic diameter ≤200 nm) consisting primarily of traffic-related emissions was collected from an urban area in Los Angeles. Male C57BL/6J mice were exposed to either re-aerosolized nPM or filtered air for 5 hours/day, 3 days/week, for 10 weeks (150 hours; n = 18/group). Microglia were characterized by immunohistochemical double staining of ionized calcium-binding protein-1 (Iba-1) with inducible nitric oxide synthase (iNOS) to identify pro-inflammatory cells, and Iba-1 with arginase-1 (Arg) to identify anti-inflammatory/ homeostatic cells. Myelin injury was assessed by degraded myelin basic protein (dMBP). Oligodendrocyte cell counts were evaluated by oligodendrocyte transcription factor 2 (Olig2). Axonal injury was assessed by axonal neurofilament marker SMI-312. iNOS-expressing microglia were significantly increased in the corpus callosum of mice exposed to nPM when compared to those exposed to filtered air (2.2 fold increase; p<0.05). This was accompanied by an increase in dMBP (1.4 fold increase; p<0.05) immunofluorescent density, a decrease in oligodendrocyte cell counts (1.16 fold decrease; p<0.05), and a decrease in neurofilament SMI-312 (1.13 fold decrease; p<0.05) immunofluorescent density. Exposure to nPM results in increased inflammatory microglia, white matter injury, and axonal degradation in the corpus callosum of adult male mice. iNOS-expressing microglia release cytokines and reactive oxygen/ nitrogen species which may further contribute to the white matter damage observed in this model.
Collapse
Affiliation(s)
- Michelle Connor
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Krista Lamorie-Foote
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
| | - Kristina Shkirkova
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
| | - Hans Baertsch
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, United States of America
| | - Todd E. Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - William J. Mack
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, United States of America
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
13
|
Aggregated Tau-PHF6 (VQIVYK) Potentiates NLRP3 Inflammasome Expression and Autophagy in Human Microglial Cells. Cells 2021; 10:cells10071652. [PMID: 34209408 PMCID: PMC8304967 DOI: 10.3390/cells10071652] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Intra-neuronal misfolding of monomeric tau protein to toxic β-sheet rich neurofibrillary tangles is a hallmark of Alzheimer’s disease (AD). Tau pathology correlates not only with progressive dementia but also with microglia-mediated inflammation in AD. Amyloid-beta (Aβ), another pathogenic peptide involved in AD, has been shown to activate NLRP3 inflammasome (NOD-like receptor family, pyrin domain containing 3), triggering the secretion of proinflammatory interleukin-1β (IL1β) and interleukin-18 (IL18). However, the effect of tau protein on microglia concerning inflammasome activation, microglial polarization, and autophagy is poorly understood. In this study, human microglial cells (HMC3) were stimulated with the unaggregated and aggregated forms of the tau-derived PHF6 peptide (VQIVYK). Modulation of NLRP3 inflammasome was examined by qRT-PCR, immunocytochemistry, and Western blot. We demonstrate that fibrillar aggregates of VQIVYK upregulated the NLRP3 expression at both mRNA and protein levels in a dose- and time-dependent manner, leading to increased expression of IL1β and IL18 in HMC3 cells. Aggregated PHF6-peptide also activated other related inflammation and microglial polarization markers. Furthermore, we also report a time-dependent effect of the aggregated PHF6 on BECN1 (Beclin-1) expression and autophagy. Overall, the PHF6 model system-based study may help to better understand the complex interconnections between Alzheimer’s PHF6 peptide aggregation and microglial inflammation, polarization, and autophagy.
Collapse
|
14
|
17-β Estradiol Rescued Immature Rat Brain against Glutamate-Induced Oxidative Stress and Neurodegeneration via Regulating Nrf2/HO-1 and MAP-Kinase Signaling Pathway. Antioxidants (Basel) 2021; 10:antiox10060892. [PMID: 34206065 PMCID: PMC8229583 DOI: 10.3390/antiox10060892] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
Dysregulated glutamate signaling, leading to neuronal excitotoxicity and death, has been associated with neurodegenerative pathologies. 17β-estradiol (E2) is a human steroid hormone having a role in reproduction, sexual maturation, brain health and biological activities. The study aimed to explain the neuroprotective role of E2 against glutamate-induced ROS production, MAP kinase-dependent neuroinflammation, synaptic dysfunction and neurodegeneration in the cortex and hippocampus of postnatal day 7 rat brain. Biochemical and immunofluorescence analyses were applied. Our results showed that a single subcutaneous injection of glutamate (10 mg/kg) induced brain oxidative stress after 4 h by disturbing the homeostasis of glutathione (GSH) and revealed an upsurge in ROS and LPO levels and downregulated the expression of Nrf2 and HO-1 antioxidant protein. The glutamate-exposed P7 pups illustrated increased phosphorylation of stress-activated c-Jun N-terminal kinase (JNK) and p38 kinase (p38) and downregulated expression of P-Erk1/2. This was accompanied by pathological neuroinflammation as revealed by enhanced gliosis with upregulated expression of GFAP and Iba-1, and the activation of proinflammatory cytokines (TNF-α) in glutamate-injected P7 pups. Moreover, exogenous glutamate also reduced the expression of synaptic markers (PSD-95, SYP) and induced apoptotic neurodegeneration in the cortical and hippocampal regions by dysregulating the expression of Bax, Bcl-2 and caspase-3 in the developing rat brain. On the contrary, co-treatment of E2 (10 mg/kg) with glutamate significantly abrogated brain neuroinflammation, neurodegeneration and synapse loss by alleviating brain oxidative stress by upregulating the Nrf2/HO-1 antioxidant pathway and by deactivating pro-apoptotic P-JNK/P-p38 and activation of pro-survival P-Erk1/2 MAP kinase pathways. In brief, the data demonstrate the neuroprotective role of E2 against glutamate excitotoxicity-induced neurodegeneration. The study also encourages future studies investigating if E2 may be a potent neuroprotective and neurotherapeutic agent in different neurodegenerative diseases.
Collapse
|
15
|
Marques DA, Gargaglioni LH, Joseph V, Bretzner F, Bícego KC, Fournier S, Kinkead R. Impact of ovariectomy and CO 2 inhalation on microglia morphology in select brainstem and hypothalamic areas regulating breathing in female rats. Brain Res 2021; 1756:147276. [PMID: 33422531 DOI: 10.1016/j.brainres.2021.147276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 01/02/2021] [Indexed: 11/17/2022]
Abstract
The neural network that regulates breathing shows a significant sexual dimorphism. Ovarian hormones contribute to this distinction as, in rats, ovariectomy reduces the ventilatory response to CO2. Microglia are neuroimmune cells that are sensitive to neuroendocrine changes in their environment. When reacting to challenging conditions, these cells show changes in their morphology that reflect an augmented capacity for producing pro- and anti-inflammatory cytokines. Based on evidence suggesting that microglia contribute to sex-based differences in reflexive responses to hypercapnia, we hypothesized that ovariectomy and hypercapnia promote microglial reactivity in selected brain areas that regulate breathing. We used ionized calcium-binding-adapter molecule-1 (Iba1) immunolabeling to compare the density and morphology of microglia in the locus coeruleus (LC), the caudal medullary raphe, the caudal part of the nucleus of the tractus solitarius (cNTS), and the paraventricular nucleus of the hypothalamus (PVN). Tissue was obtained from SHAM (metaestrus) female rats or following ovariectomy. Rats were exposed to normocapnia or hypercapnia (5% CO2, 20 min). Ovariectomy and hypercapnia did not affect microglial density in any of the structures studied. Ovariectomy promoted a reactive phenotype in the cNTS and LC, as indicated by a larger morphological index. In these structures, hypercapnia had a relatively modest opposing effect; the medullary raphe or the PVN were not affected. We conclude that ovarian hormones attenuate microglial reactivity in CO2/H+ sensing structures. These data suggest that microglia may contribute to neurological diseases in which anomalies of respiratory control are associated with cyclic fluctuations of ovarian hormones or menopause.
Collapse
Affiliation(s)
- Danuzia A Marques
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada.
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Vincent Joseph
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Frédéric Bretzner
- Département de Psychiatrie et Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Stéphanie Fournier
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada
| |
Collapse
|
16
|
Aryanpour R, Zibara K, Pasbakhsh P, Jame'ei SB, Namjoo Z, Ghanbari A, Mahmoudi R, Amani S, Kashani IR. 17β-Estradiol Reduces Demyelination in Cuprizone-fed Mice by Promoting M2 Microglia Polarity and Regulating NLRP3 Inflammasome. Neuroscience 2021; 463:116-127. [PMID: 33794337 DOI: 10.1016/j.neuroscience.2021.03.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/18/2021] [Accepted: 03/21/2021] [Indexed: 11/26/2022]
Abstract
Estrogen produces a beneficial role in animal models of multiple sclerosis (MS). The effect of 17β-estradiol therapy on microglia polarization and neuroinflammation in the corpus callosum of the cuprizone-induced demyelination model has not been elucidated. In this study, mice were given 0.2% cuprizone (CPZ) for 5 weeks to induce demyelination during which they received 50 ng of 17β-estradiol (EST), injected subcutaneously in the neck region, twice weekly. Data revealed that treatment with 17β-estradiol therapy (CPZ+EST) improved neurological behavioral deficits, displayed by a significant reduction in escape latencies, in comparison to untreated CPZ mice. Also, administration of 17β-estradiol caused a decrease in demyelination levels and axonal injury, as demonstrated by staining with Luxol fast blue, immunofluorescence to myelin basic protein, and transmission electron microscopy analysis. In addition, at the transcriptional level in the brain, mice treated with 17β-estradiol (CPZ+EST) showed a decrease in the levels of M1-assosicted microglia markers (CD86, iNOS and MHC-II) whereas M2-associated genes (Arg-1, CD206 and Trem-2) were increased, compared to CPZ mice. Moreover, administration of 17β-estradiol resulted in a significant reduction (∼3-fold) in transcript levels of NLRP3 inflammasome and its downstream product IL-18, compared to controls. In summary, this study demonstrated for the first time that exogenous 17β-estradiol therapy robustly leads to the reduction of M1 phenotype, stimulation of polarized M2 microglia, and repression of NLRP3 inflammasome in the corpus callosum of CPZ demyelination model of MS. The positive effects of 17β-estradiol on microglia and inflammasome seems to facilitate and accelerate the remyelination process.
Collapse
Affiliation(s)
- Roya Aryanpour
- Department of Anatomy, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zeinab Namjoo
- Department of Anatomical Science, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amir Ghanbari
- Cell and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Reza Mahmoudi
- Cell and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Showan Amani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Roosen K, Scheld M, Mandzhalova M, Clarner T, Beyer C, Zendedel A. CXCL12 inhibits inflammasome activation in LPS-stimulated BV2 cells. Brain Res 2021; 1763:147446. [PMID: 33766517 DOI: 10.1016/j.brainres.2021.147446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 10/21/2022]
Abstract
The activation of the CXCL12-CXCR4 signaling axis is implicated in the regulation of cell survival, proliferation, and mobilization of bone marrow stem cells into the injured site. We have shown in a previous study that intrathecal administration of CXCL12 reduces spinal cord tissue damage and neuroinflammation and provides functional improvement by reducing inflammasome activity and local inflammatory processes in an experimental spinal cord injury (SCI) rat model. Here, we aimed at investigating whether these neuroprotective effects rely on the control of CXCL12 signaling on microglial activation as microglia cells are known to be the primary immune cells of the brain. LPS induced the expression of the inflammasome components NLRP3, NLRC4 and ASC, the secretion of the cytokines IL-1b and IL-18 and the activation of caspase-1 protease in BV2 cells. Pre-treatment with CXCL12 significantly reduced LPS-induced IL-1b/IL-18 secretion and inflammasome induction. Our results also showed that CXCL12 can suppress caspase-1 activity, which leads to a decrease of SCI-related induction of active IL-1b.
Collapse
Affiliation(s)
- Kenza Roosen
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Miriam Scheld
- Anatomy and Cell Biology, University of Augsburg, 86159 Augsburg, Germany
| | | | - Tim Clarner
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
18
|
Voelz C, Habib P, Köberlein S, Beyer C, Slowik A. Alteration of miRNA Biogenesis Regulating Proteins in the Human Microglial Cell Line HMC-3 After Ischemic Stress. Mol Neurobiol 2020; 58:1535-1549. [PMID: 33210205 DOI: 10.1007/s12035-020-02210-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNA) are small noncoding sequences that control apoptosis, proliferation, and neuroinflammatory pathways in microglia cells. The expression of distinct miRNAs is altered after ischemia in the brain. Only minor information is available about the biogenesis and maturation of miRNAs after ischemia. We aimed at examining the impact of oxygen-glucose deprivation (OGD) and hydrogen peroxide (H2O2)-induced stress on the expression of miRNA regulating proteins such as DROSHA, DGCR8, XPO5, DICER, TARBP2, and AGO2 in the cultured human microglial cell line HMC-3 (human microglial cell line clone 3). OGD duration of 2.5 h or H2O2 stimulation at a concentration of 100 μM for 24 h resulted in a marked increase of the hypoxia sensor hypoxia-inducible factor1-α in HMC-3 cells. These treatments also led to an upregulation of DROSHA, DICER1, and AGO2 detected by semiquantitative real-time PCR (qrtPCR). XPO5 and TARBP2 were only upregulated after stimulation with H2O2, while DGCR8 responded only to OGD. We found elevated DICER1, DROSHA, and AGO2 protein levels by western blot and immunohistochemistry staining. Interestingly, the latter also exposed a colocalization of AGO2 with stress granules (G3BP1) after OGD. Our data indicate that DICER, DROSHA, and AGO2 are induced in microglial cells under hypoxia-like conditions. It might be speculated that their inductions might increase the miRNA synthesis rate. Future studies should investigate this correlation to determine which miRNAs are preferably expressed by microglia cells after ischemia and which functions they could exert.
Collapse
Affiliation(s)
- Clara Voelz
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Stefan Köberlein
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
- JARA-Brain, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
19
|
Gamache J, Yun Y, Chiba-Falek O. Sex-dependent effect of APOE on Alzheimer's disease and other age-related neurodegenerative disorders. Dis Model Mech 2020; 13:dmm045211. [PMID: 32859588 PMCID: PMC7473656 DOI: 10.1242/dmm.045211] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The importance of apolipoprotein E (APOE) in late-onset Alzheimer's disease (LOAD) has been firmly established, but the mechanisms through which it exerts its pathogenic effects remain elusive. In addition, the sex-dependent effects of APOE on LOAD risk and endophenotypes have yet to be explained. In this Review, we revisit the different aspects of APOE involvement in neurodegeneration and neurological diseases, with particular attention to sex differences in the contribution of APOE to LOAD susceptibility. We discuss the role of APOE in a broader range of age-related neurodegenerative diseases, and summarize the biological factors linking APOE to sex hormones, drawing on supportive findings from rodent models to identify major mechanistic themes underlying the exacerbation of LOAD-associated neurodegeneration and pathology in the female brain. Additionally, we list sex-by-genotype interactions identified across neurodegenerative diseases, proposing APOE variants as a shared etiology for sex differences in the manifestation of these diseases. Finally, we present recent advancements in 'omics' technologies, which provide a new platform for more in-depth investigations of how dysregulation of this gene affects the development and progression of neurodegenerative diseases. Collectively, the evidence summarized in this Review highlights the interplay between APOE and sex as a key factor in the etiology of LOAD and other age-related neurodegenerative diseases. We emphasize the importance of careful examination of sex as a contributing factor in studying the underpinning genetics of neurodegenerative diseases in general, but particularly for LOAD.
Collapse
Affiliation(s)
- Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Young Yun
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
20
|
Habib P, Harms J, Zendedel A, Beyer C, Slowik A. Gonadal Hormones E2 and P Mitigate Cerebral Ischemia-Induced Upregulation of the AIM2 and NLRC4 Inflammasomes in Rats. Int J Mol Sci 2020; 21:ijms21134795. [PMID: 32645874 PMCID: PMC7370209 DOI: 10.3390/ijms21134795] [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: 06/15/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Acute ischemic stroke (AIS) is a devastating neurological condition with a lack of neuroprotective therapeutic options, despite the reperfusion modalities thrombolysis and thrombectomy. Post-ischemic brain damage is aggravated by an excessive inflammatory cascade involving the activation and regulation of the pro-inflammatory cytokines IL-1β and IL-18 by inflammasomes. However, the role of AIM2 and NLRC4 inflammasomes and the influence of the neuroprotective steroids 17β-estradiol (E2) and progesterone (P) on their regulation after ischemic stroke have not yet been conclusively elucidated. To address the latter, we subjected a total of 65 rats to 1 h of transient Middle Cerebral Artery occlusion (tMCAO) followed by a reperfusion period of 72 h. Moreover, we evaluated the expression and regulation of AIM2 and NLRC4 in glial single-cell cultures (astroglia and microglia) after oxygen–glucose deprivation (OGD). The administration of E2 and P decreased both infarct sizes and neurological impairments after cerebral ischemia in rats. We detected a time-dependent elevation of gene and protein levels (Western Blot/immunohistochemistry) of the AIM2 and NLRC4 inflammasomes in the post-ischemic brains. E2 or P selectively mitigated the stroke-induced increase of AIM2 and NLRC4. While both inflammasomes seemed to be exclusively abundant in neurons under physiological and ischemic conditions in vivo, single-cell cultures of cortical astrocytes and microglia equally expressed both inflammasomes. In line with the in vivo data, E and P selectively reduced AIM2 and NLRC4 in primary cortical astrocytes and microglial cells after OGD. In conclusion, the post-ischemic elevation of AIM2 and NLRC4 and their down-regulation by E2 and P may shed more light on the anti-inflammatory effects of both gonadal hormones after stroke.
Collapse
Affiliation(s)
- Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany;
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Julie Harms
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (J.H.); (A.Z.); (C.B.)
| | - Adib Zendedel
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (J.H.); (A.Z.); (C.B.)
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (J.H.); (A.Z.); (C.B.)
- JARA Brain, RWTH Aachen University, 52074 Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (J.H.); (A.Z.); (C.B.)
- Correspondence: ; Tel.: +49-241-80-89112
| |
Collapse
|
21
|
Acosta-Martínez M. Shaping Microglial Phenotypes Through Estrogen Receptors: Relevance to Sex-Specific Neuroinflammatory Responses to Brain Injury and Disease. J Pharmacol Exp Ther 2020; 375:223-236. [DOI: 10.1124/jpet.119.264598] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
|
22
|
Microglial-specific depletion of TAK1 is neuroprotective in the acute phase after ischemic stroke. J Mol Med (Berl) 2020; 98:833-847. [PMID: 32382778 PMCID: PMC7297861 DOI: 10.1007/s00109-020-01916-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/01/2020] [Accepted: 04/22/2020] [Indexed: 12/24/2022]
Abstract
Abstract Transforming growth factor-β-activated kinase 1 (TAK1) is upregulated after cerebral ischemia and contributes to an aggravation of brain injury. TAK1 acts as a key regulator of NF-ΚB and the MAP kinases JNK and p38 and modulates post-ischemic neuroinflammation and apoptosis. Microglia are the main TAK1-expressing immunocompetent cells of the brain. However, little is known about the function and regulation of microglial TAK1 after cerebral ischemia. Tamoxifen-dependent conditional depletion of TAK1 in microglial cells was induced in Cx3cr1creER-Tak1fl/fl mice. The creER-negative Tak1fl/fl mice and vehicle-treated (corn oil) mice served as control groups. A transient intraluminal middle cerebral artery occlusion of 30 min followed by 6 h and 72 h of reperfusion was performed in male mice. Oxygen-glucose-deprivation (OGD) was performed with primary cortical glial cell cultures to examine the effect of microglial-specific and general (5Z-7-Oxozeaenol) TAK1 inhibition after different reperfusion times (1 h, 6 h, and 72 h). Cx3cr1creER-Tak1fl/fl mice showed reduced infarct sizes and improved neurological outcomes compared to the control group. The mRNA and protein levels of pro-inflammatory Il1b/IL-1β and Tnf/TNF-α in the peri-infarct zones of microglial-specific TAK1-depleted mice were significantly reduced. Furthermore, TAK1 depletion in vitro led to reduced cell death rates after OGD. Moreover, hypoxia-mediated activation of TAK1 and its downstream signalling proteins, JNK and p38, were dampened by microglial TAK1 depletion. In contrast, 5Z-7-Oxozeaenol-induced pharmacological inhibition of TAK1 completely diminished MAPK-signalling including the kinases JNK and p38 in all cells. Microglial TAK1 depletion abrogates post-ischemic neuroinflammation and apoptosis in the acute phase, hence might be considered as a potential target in the treatment of cerebral hypoxia. Key messages TAK1 is activated after cerebral ischemia and induces MAP kinases p38 and JNK. Activated TAK1 increases apoptosis rate and the level pro-inflammatory cytokines IL-1β and TNF-α. Microglial cells seem to be the main source of TAK1-mediated post-ischemic neuroinflammation. Microglial-specific TAK1-depletion mediates sustainable neuroprotective effects, which might be superior to global TAK1 inhibition.
Electronic supplementary material The online version of this article (10.1007/s00109-020-01916-9) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Schölwer I, Habib P, Voelz C, Rolfes L, Beyer C, Slowik A. NLRP3 Depletion Fails to Mitigate Inflammation but Restores Diminished Phagocytosis in BV-2 Cells After In Vitro Hypoxia. Mol Neurobiol 2020; 57:2588-2599. [PMID: 32239449 DOI: 10.1007/s12035-020-01909-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/23/2020] [Indexed: 12/30/2022]
Abstract
Post-hypoxic/ischemic neuroinflammation is selectively driven by sterile inflammation, which implies the interplay of brain-intrinsic immune cells with other neural cells and immigrated peripheral immune cells. The resultant inflammatory cascade evolves extra- and intracellular pathogen and danger-associated receptors. The latter interacts with multiprotein complexes termed inflammasomes. The NLRP3 inflammasome is one of the best-described inflammasomes. However, its impact on post-ischemic neuroinflammation and its role in neuroprotection after ischemic stroke are still under debate. Microglial cells are known to be the main source of neuroinflammation; hence, we depleted NLRP3 in BV-2 microglial cells using shRNA to investigate its role in IL-1β maturation and phagocytosis after hypoxia (oxygen-glucose-deprivation (OGD)). We also examined the expression profiles of other inflammasomes (NLRC4, AIM2, ASC) and caspase-1 activity after OGD. OGD triggered caspase-1 activity and increased IL-1β secretion in BV-2 cells with no alteration after NLRP3 depletion. The expression of the AIM2 inflammasome was significantly higher after OGD in NLRP3-depleted cells, whereas NLRC4 was unaltered in all groups. Interestingly, OGD induced a complete inactivation of phagocytic activity in wild-type cells, while in NLRP3-depleted BV-2, this inactivity was restored after hypoxia. Our findings indicate a minor role of NLRP3 in the inflammatory response after hypoxic/ischemic stimulus. However, NLRP3 seems to play a pivotal role in the regulation of post-ischemic phagocytosis. This might be a prerequisite for the putative neuroprotective effect.
Collapse
Affiliation(s)
- Isabelle Schölwer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Clara Voelz
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Leoni Rolfes
- Neurology Clinic and Institute for Translational Neurology, University of Muenster, Münster, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-Brain, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
24
|
Habib P, Stamm AS, Schulz JB, Reich A, Slowik A, Capellmann S, Huber M, Wilhelm T. EPO and TMBIM3/GRINA Promote the Activation of the Adaptive Arm and Counteract the Terminal Arm of the Unfolded Protein Response after Murine Transient Cerebral Ischemia. Int J Mol Sci 2019; 20:ijms20215421. [PMID: 31683519 PMCID: PMC6862264 DOI: 10.3390/ijms20215421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke is known to cause the accumulation of misfolded proteins and loss of calcium homeostasis leading to impairment of endoplasmic reticulum (ER) function. The unfolded protein response (UPR) is an ER-located and cytoprotective pathway that aims to resolve ER stress. Transmembrane BAX inhibitor-1 motif-containing (TMBIM) protein family member TMBIM3/GRINA is highly expressed in the brain and mostly located at the ER membrane suppressing ER calcium release by inositol-1,4,5-trisphosphate receptors. GRINA confers neuroprotection and is regulated by erythropoietin (EPO) after murine cerebral ischemia. However, the role of GRINA and the impact of EPO treatment on the post-ischemic UPR have not been elucidated yet. We subjected GRINA-deficient (Grina−/−) and wildtype mice to transient (30 min) middle cerebral artery occlusion (tMCAo) followed by 6 h or 72 h of reperfusion. We administered EPO or saline 0, 24 and 48 h after tMCAo/sham surgery. Oxygen–glucose deprivation (OGD) and pharmacological stimulation of the UPR using Tunicamycin and Thapsigargin were carried out in primary murine cortical mixed cell cultures. Treatment with the PERK-inhibitor GSK-2606414, IRE1a-RNase-inhibitor STF-083010 and EPO was performed 1 h prior to either 1 h, 2 h or 3 h of OGD. We found earlier and larger infarct demarcations in Grina−/− mice compared to wildtype mice, which was accompanied by a worse neurological outcome and an abolishment of EPO-mediated neuroprotection after ischemic stroke. In addition, GRINA-deficiency increased apoptosis and the activation of the corresponding PERK arm of the UPR after stroke. EPO enhanced the post-ischemic activation of pro-survival IRE1a and counteracted the pro-apoptotic PERK branch of the UPR. Both EPO and the PERK-inhibitor GSK-2606414 reduced cell death and regulated Grina mRNA levels after OGD. In conclusion, GRINA plays a crucial role in post-ischemic UPR and the use of both GSK-2606414 and EPO might lead to neuroprotection.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Adenine/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Cells, Cultured
- Endoplasmic Reticulum Stress/drug effects
- Endoplasmic Reticulum Stress/genetics
- Erythropoietin/pharmacology
- Glucose/metabolism
- Indoles/pharmacology
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/prevention & control
- Ischemic Attack, Transient/genetics
- Ischemic Attack, Transient/metabolism
- Ischemic Attack, Transient/prevention & control
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuroprotective Agents/pharmacology
- Oxygen/metabolism
- Sulfonamides/pharmacology
- Thapsigargin/pharmacology
- Thiophenes/pharmacology
- Tunicamycin/pharmacology
- Unfolded Protein Response/drug effects
- Unfolded Protein Response/genetics
Collapse
Affiliation(s)
- Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| | - Ann-Sophie Stamm
- Department of Neurology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| | - Joerg B Schulz
- Department of Neurology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
- JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbAnd RWTH Aachen University, 52074 Aachen, Germany.
| | - Arno Reich
- Department of Neurology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| | - Sandro Capellmann
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
25
|
Habib P, Stamm AS, Zeyen T, Noristani R, Slowik A, Beyer C, Wilhelm T, Huber M, Komnig D, Schulz JB, Reich A. EPO regulates neuroprotective Transmembrane BAX Inhibitor-1 Motif-containing (TMBIM) family members GRINA and FAIM2 after cerebral ischemia-reperfusion injury. Exp Neurol 2019; 320:112978. [PMID: 31211943 DOI: 10.1016/j.expneurol.2019.112978] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/03/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Transmembrane BAX Inhibitor-1 Motif-containing (TMBIM) family members exert inhibitory activities in apoptosis and necroptosis. FAIM2 (TMBIM-2) is neuroprotective against murine focal ischemia and is regulated by erythropoietin (EPO). Similar to FAIM2, GRINA (TMBIM-3) is predominantly expressed in the brain. The role of GRINA in transient brain ischemia, its potential synergistic effects with FAIM2 and its regulation by EPO treatment were assessed. METHODS We performed transient (30 min) middle cerebral artery occlusion (tMCAo) followed by 72 h of reperfusion in GRINA-deficient (GRINA-/-), FAIM2-deficient (FAIM2-/-), double-deficient (GRINA-/-FAIM2-/-) and wildtype littermates (WT) mice. We administered EPO or saline 0, 24 and 48 h after tMCAo. We subjected primary murine cortical neurons (pMCN) of all mouse strains to oxygen-glucose deprivation (OGD) after GRINA and/or FAIM2 gene transfection. RESULTS Compared to wildtype controls GRINA deficiency led to a similar increase in infarct volumes as FAIM2 deficiency (p < .01). We observed the highest neurological deficits and largest infarct sizes in double-deficient mice. EPO administration upregulated GRINA and FAIM2 mRNA levels in wildtype littermates. EPO decreased infarct sizes and abrogated neurological impairments in wildtype controls. GRINA and/or FAIM2 deficient mice showed increased expression levels of cleaved-caspase 3 and of pro-apoptotic BAX mRNA. Further, caspase 8 was upregulated in FAIM2-/- and caspase 9 in GRINA-/- mice. Overexpression of GRINA and FAIM2 in wildtype and in double deficient pMCN decreased cell death rate after OGD. CONCLUSIONS GRINA and FAIM2 are highly expressed in the brain and convey EPO-mediated neuroprotection after ischemic stroke involving different caspases.
Collapse
Affiliation(s)
- Pardes Habib
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany.
| | - Ann-Sophie Stamm
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Thomas Zeyen
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Rozina Noristani
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical School, RWTH Aachen University, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical School, RWTH Aachen University, Aachen, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Daniel Komnig
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Jörg B Schulz
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany; JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, Medical School, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
26
|
Aftabizadeh M, Tatarek-Nossol M, Andreetto E, El Bounkari O, Kipp M, Beyer C, Latz E, Bernhagen J, Kapurniotu A. Blocking Inflammasome Activation Caused by β-Amyloid Peptide (Aβ) and Islet Amyloid Polypeptide (IAPP) through an IAPP Mimic. ACS Chem Neurosci 2019; 10:3703-3717. [PMID: 31295403 DOI: 10.1021/acschemneuro.9b00260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inflammation in the brain and pancreas is linked to cell degeneration and pathogenesis of both Alzheimer's disease (AD) and type 2 diabetes (T2D). Inflammatory cascades in both tissues are triggered by the uptake of β-amyloid peptide (Aβ) or islet amyloid polypeptide (IAPP) aggregates by microglial cells (AD) or macrophages (T2D) and their insufficient lysosomal degradation. This results in lysosomal damage, caspase-1/NLRP3 inflammasome activation and release of interleukin-1β (IL-1β), a key proinflammatory cytokine in both diseases. Here we show that the inflammatory processes mediated by Aβ and IAPP aggregates in microglial cells and macrophages are blocked by IAPP-GI, a nonamyloidogenic IAPP mimic, which forms high-affinity soluble and nonfibrillar hetero-oligomers with both polypeptides. In contrast to fibrillar Aβ aggregates, nonfibrillar Aβ/IAPP-GI or Aβ/IAPP hetero-oligomers become rapidly internalized by microglial cells and targeted to lysosomes where Aβ is fully degraded. Internalization occurs via IAPP receptor-mediated endocytosis. Moreover, in contrast to IAPP aggregates, IAPP/IAPP-GI hetero-oligomers become rapidly internalized and degraded in the lysosomal compartments of macrophages. Our findings uncover a previously unknown function for the IAPP/Aβ cross-amyloid interaction and suggest that conversion of Aβ or IAPP into lysosome-targeted and easily degradable hetero-oligomers by heteroassociation with IAPP mimics could become a promising approach to specifically prevent amyloid-mediated inflammation in AD, T2D, or both diseases.
Collapse
Affiliation(s)
- Maryam Aftabizadeh
- Division of Peptide Biochemistry, Technische Universität München, Emil-Erlenmeyer-Forum 5, D-85354 Freising, Germany
- Cancer Immunotherapeutics and Tumor Immunology, City of Hope Medical Center Duarte, 1500 East Duarte Road, Duarte, California 91010, United States
| | | | - Erika Andreetto
- Division of Peptide Biochemistry, Technische Universität München, Emil-Erlenmeyer-Forum 5, D-85354 Freising, Germany
| | - Omar El Bounkari
- Chair of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany
| | | | - Eicke Latz
- Institute of Innate Immunity, University of Bonn, Biomedical Center, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
- Division of Infectious Diseases & Immunology, University of Massachusetts Medical School, 364 Plantation St., Worcester, Massachusetts 01605, United States
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, Technische Universität München, Emil-Erlenmeyer-Forum 5, D-85354 Freising, Germany
| |
Collapse
|
27
|
Azedi F, Mehrpour M, Talebi S, Zendedel A, Kazemnejad S, Mousavizadeh K, Beyer C, Zarnani AH, Joghataei MT. Melatonin regulates neuroinflammation ischemic stroke damage through interactions with microglia in reperfusion phase. Brain Res 2019; 1723:146401. [PMID: 31445031 DOI: 10.1016/j.brainres.2019.146401] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022]
Abstract
Even today, ischemic stroke is a major cause of death and disabilities because of its high incidence, limited treatments and poor understanding of the pathophysiology of ischemia/reperfusion, neuroinflammation and secondary injuries following ischemic stroke. The function of microglia as a part of the immune system of the brain following ischemic stroke can be destructive or protective. Recent surveys indicate that melatonin, a strong antioxidant agent, has receptors on microglial cells and can regulate them to protective form; yet, more findings are required for better understanding of this mechanism, particularly in the reperfusion phase. In this study, we initially aimed to evaluate the therapeutic efficacy of melatonin intra-arterially and to clarify the underlying mechanisms. After that by using an in vitro approach, we evaluated the protective effects of melatonin on microglial cells following the hypoxia condition. Our results proved that a single dose of melatonin at the beginning of reperfusion phase improved structural and behavioral outcomes. Melatonin increased NeuN and decreased GFAP, Iba1 and active caspase-3 at protein level. Furthermore, melatonin elevated BDNF, MAP2, HSPA1A and reduced VEGF at mRNA level. We also showed that melatonin receptor 1B highly expressed in microglial cells after 3 h hypoxia. Besides, melatonin increased the ratio of TREM2/iNOS as a marker of the most protective form of microglia (M2). In summary, our data suggest that melatonin has the possibility to serve as targeting microglial action for preventing secondary injury of reperfusion phase after ischemic stroke.
Collapse
Affiliation(s)
- Fereshteh Azedi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Mehrpour
- Department of Neurology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Talebi
- Department of Medical Genetics, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Somaieh Kazemnejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Kazem Mousavizadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Butturini E, Boriero D, Carcereri de Prati A, Mariotto S. STAT1 drives M1 microglia activation and neuroinflammation under hypoxia. Arch Biochem Biophys 2019; 669:22-30. [PMID: 31121156 DOI: 10.1016/j.abb.2019.05.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/07/2019] [Accepted: 05/15/2019] [Indexed: 12/25/2022]
Abstract
Microglia are resident immune cells that act as the first active defence in the central nervous system. These cells constantly monitor the tissue microenvironment and rapidly react in response to hypoxia, infection and injuries. Hypoxia in the brain has been detected in several neurodegenerative disorders such as Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. Hypoxic conditions activate microglia cells towards M1 phenotype resulting in oxidative stress and the release of pro-inflammatory cytokines. Recently, we have demonstrated that oxidative stress induces S-glutathionylation of the STAT1 and hyper-activates its signaling in microglia BV2 cells pointing out the importance of this transcription factor in neuroinflammation. In this paper we analyse the cellular mechanisms that drive M1 microglia activation in BV2 cells in response to hypoxia correlating it to STAT1 activation. The analysis of the molecular mechanism of STAT1 signaling reveals that hypoxia generates oxidative stress and induces both phosphorylation and S-glutathionylation of STAT1 that are responsible of its aberrant activation. The silencing of STAT1 protein expression counteracts hypoxia-M1 microglia phenotype suggesting the strong link between hypoxia-STAT1 and STAT1-microglia activation.
Collapse
Affiliation(s)
- Elena Butturini
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy.
| | - Diana Boriero
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| | - Alessandra Carcereri de Prati
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| | - Sofia Mariotto
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| |
Collapse
|
29
|
Butler MJ, Eckel LA. Eating as a motivated behavior: modulatory effect of high fat diets on energy homeostasis, reward processing and neuroinflammation. Integr Zool 2019; 13:673-686. [PMID: 29851251 DOI: 10.1111/1749-4877.12340] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Eating is a basic motivated behavior that provides fuel for the body and supports brain function. To ensure survival, the brain's feeding circuits are tuned to monitor peripheral energy balance and promote food-seeking behavior when energy stores are low. The brain's bias toward a positive energy state, which is necessary to ensure adequate nutrition during times of food scarcity, is evolutionarily conserved across mammalian species and is likely to drive overeating in the presence of a palatable, energy-dense diet. Animal models of diet-induced overeating have played a vital role in investigating how the drive to consume palatable food may override the homeostatic processes that serve to maintain energy balance. These animal models have provided valuable insights into the neurobiological mechanisms underlying homeostatic and non-homeostatic eating, motivation and food reward, and the development of obesity and related comorbidities. Here, we provide a brief review of this literature and discuss how diet-induced inflammation in the central nervous system impacts the neural control of food intake and regulation of body weight. The connection between diet and the immune system provides an exciting new direction for the study of ingestive behavior and the pathophysiology of obesity.
Collapse
Affiliation(s)
- Michael J Butler
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Lisa A Eckel
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
30
|
Slowik A, Lammerding L, Zendedel A, Habib P, Beyer C. Impact of steroid hormones E2 and P on the NLRP3/ASC/Casp1 axis in primary mouse astroglia and BV-2 cells after in vitro hypoxia. J Steroid Biochem Mol Biol 2018; 183:18-26. [PMID: 29772377 DOI: 10.1016/j.jsbmb.2018.05.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/02/2018] [Accepted: 05/13/2018] [Indexed: 11/29/2022]
Abstract
Clinical and animal model studies have demonstrated the neuroprotective and anti-inflammatory effects of 17beta-estradiol (E2) and progesterone (P) in different disease models of the central nervous system (CNS) including ischemic stroke. Inflammasomes are involved in the interleukin-1 beta (IL1beta) maturation, in particular, NLRP3, the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) and the active caspase-1 (Casp1) form. Recently, we showed that administration of E2 or P selectively regulated these components after experimental ischemic stroke in rats. Therefore, we investigated the impact of E2 and P on the NLRP3/ASC/Casp1 axis in the murine microglia-like cell line BV-2 cells and primary astrocytes after short-term in vitro hypoxia. The inflammatory cytokine IL1beta but not IL18 was increased after short-term hypoxia in astroglia and BV-2 cells. The same applied to NLPR3 and ASC. Casp1 activity was also elevated in astroglia and BV-2 cells after hypoxia. The administration of E2 or P selectively dampened IL1beta, ASC and NLRP3 expression mainly in BV-2 cells. Both steroid hormones failed to reduce Casp1 activity after hypoxia. We conclude that E2- and P-mediated anti-inflammatory mechanisms occur upstream of Casp1 through the regulation of NLRP3 and its adaptor ASC.
Collapse
Affiliation(s)
- Alexander Slowik
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany.
| | - Leoni Lammerding
- Department of Neurology, University Hospital of Münster, Münster, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany; Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pardes Habib
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany; JARA-Brain, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
31
|
Ebrahimi T, Rust M, Kaiser SN, Slowik A, Beyer C, Koczulla AR, Schulz JB, Habib P, Bach JP. α1-antitrypsin mitigates NLRP3-inflammasome activation in amyloid β 1-42-stimulated murine astrocytes. J Neuroinflammation 2018; 15:282. [PMID: 30261895 PMCID: PMC6158809 DOI: 10.1186/s12974-018-1319-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 09/19/2018] [Indexed: 12/28/2022] Open
Abstract
Background Neuroinflammation has an essential impact on the pathogenesis and progression of Alzheimer’s disease (AD). Mostly mediated by microglia and astrocytes, inflammatory processes lead to degeneration of neuronal cells. The NLRP3-inflammasome (NOD-like receptor family, pyrin domain containing 3) is a key component of the innate immune system and its activation results in secretion of the proinflammatory effectors interleukin-1β (IL-1β) and interleukin-18 (IL-18). Under physiological conditions, cytosolic NLRP3-inflammsome is maintained in an inactive form, not able to oligomerize. Amyloid β1–42 (Aβ1–42) triggers activation of NLRP3-inflammasome in microglia and astrocytes, inducing oligomerization and thus recruitment of proinflammatory proteases. NLRP3-inflammasome was found highly expressed in human brains diagnosed with AD. Moreover, NLRP3-deficient mice carrying mutations associated with familial AD were partially protected from deficits associated with AD. The endogenous protease inhibitor α1-antitrypsin (A1AT) is known for its anti-inflammatory and anti-apoptotic properties and thus could serve as therapeutic agent for NLRP3-inhibition. A1AT protects neurons from glutamate-induced toxicity and reduces Aβ1–42-induced inflammation in microglial cells. In this study, we investigated the effect of Aβ1–42-induced NLRP3-inflammasome upregulation in primary murine astrocytes and its regulation by A1AT. Methods Primary cortical astrocytes from BALB/c mice were stimulated with Aβ1–42 and treated with A1AT. Regulation of NLRP3-inflammasome was examined by immunocytochemistry, PCR, western blot and ELISA. Our studies included an inhibitor of NLRP3 to elucidate direct interactions between A1AT and NLRP3-inflammasome components. Results Our study revealed that A1AT reduces Aβ1–42-dependent upregulation of NLRP3 at the mRNA and protein levels. Furthermore, A1AT time-dependently mitigated the expression of caspase 1 and its cleavage product IL-1β in Aβ1–42-stimulated astrocytes. Conclusion We conclude that Aβ1–42-stimulation results in an upregulation of NLRP3, caspase 1, and its cleavage products in astrocytes. A1AT time-dependently hampers neuroinflammation by downregulation of Aβ1–42-mediated NLRP3-inflammasome expression and thus may serve as a pharmaceutical opportunity for the treatment of Alzheimer’s disease. Electronic supplementary material The online version of this article (10.1186/s12974-018-1319-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taraneh Ebrahimi
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Marcus Rust
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | | | - Alexander Slowik
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Andreas Rembert Koczulla
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Marburg, Germany
| | - Jörg B Schulz
- Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Jan Philipp Bach
- Department of Neurology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
32
|
El-Bakoush A, Olajide OA. Formononetin inhibits neuroinflammation and increases estrogen receptor beta (ERβ) protein expression in BV2 microglia. Int Immunopharmacol 2018; 61:325-337. [PMID: 29913427 DOI: 10.1016/j.intimp.2018.06.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 11/15/2022]
Abstract
Formononetin is a bioactive non-steroidal polyphenol found in a variety of plants. In this study we evaluated the effects of formononetin on neuroinflammation in LPS-stimulated BV2 microglia. Results showed that formononetin significantly reduced the production of TNF-α, IL-6 and IL-1β, nitrite and PGE2, as well as protein levels of iNOS and COX-2. Reporter gene assays showed that formononetin produced inhibition of NF-κB luciferase activity in HEK293 cells stimulated with TNF-α. Immunoblotting experiments revealed an inhibition of IKKα phosphorylation, with the resultant attenuation of phosphorylation and degradation of IκBα following LPS stimulation. Formononetin also produced an inhibition of nuclear translocation and DNA binding by NF-κB following LPS stimulation. RNAi experiments showed that transfection of BV2 microglia with ERβ siRNA resulted in the loss of anti-inflammatory action of formononetin. MTT assay and MAP2 immunoreactivity experiments showed that formononetin produced significant neuroprotective activity by preventing BV2 microglia conditioned media-induced toxicity to HT22 neurons. Investigations on the effect of formononetin on MCF7 breast cancer cells revealed that, while the compound significantly increased ER-luciferase activity, its effects on proliferation were modest. This study has established that formononetin inhibits neuroinflammation by targeting NF-κB signalling pathway in BV2 microglia, possibly through mechanisms involving ERβ. Formononetin appears to modulate ERβ in MCF7 breast cancer cells with limited proliferative effect. Formononetin could therefore serve as a chemical scaffold for the development of novel compounds which have selective neuroprotective actions in the brain.
Collapse
Affiliation(s)
- Abdelmeniem El-Bakoush
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, United Kingdom
| | - Olumayokun A Olajide
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, United Kingdom.
| |
Collapse
|
33
|
Cai B, Seong KJ, Bae SW, Chun C, Kim WJ, Jung JY. A synthetic diosgenin primary amine derivative attenuates LPS-stimulated inflammation via inhibition of NF-κB and JNK MAPK signaling in microglial BV2 cells. Int Immunopharmacol 2018; 61:204-214. [PMID: 29890414 DOI: 10.1016/j.intimp.2018.05.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 02/07/2023]
Abstract
Diosgenin, a precursor of steroid hormones in plants, is known to exhibit diverse pharmacological activities including anti-inflammatory properties. In this study, (3β, 25R)‑spirost‑5‑en‑3‑oxyl (2‑((2((2‑aminoethyl)amino)ethyl)amino)ethyl) carbamate (DGP), a new synthetic diosgenin derivative incorporating primary amine was used to investigate its anti-inflammatory effects and underlying mechanisms of action in lipopolysaccharide (LPS)-stimulated microglial BV2 cells. Pretreatment with DGP resulted in significant inhibition of nitric oxide (NO) synthesis, and down-regulation of nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-stimulated microglial BV2 cells. In addition, DGP decreased the production of reactive oxygen species (ROS) and pro-inflammatory cytokines such as interleukin (IL)-6, IL-1β, and tumor necrosis factor alpha (TNF-α). The inhibitory effects of DGP on these inflammatory mediators in LPS-stimulated microglial BV2 cells were regulated by NF-κB signaling through blocking p65 nuclear translocation and NF-κB p65/DNA binding activity. DGP also blocked the phosphorylation of c-Jun amino-terminal kinase (JNK), but not p38 kinase or extracellular signal-regulated kinases (ERK). The NF-κB inhibitor JSH-23 and JNK-specific inhibitor SP600125 significantly decreased NO production and IL-6 release in LPS-stimulated BV2 cells, respectively. The overall results demonstrate that DGP has anti-inflammatory effects on LPS-stimulated BV2 cells via inhibition of NF-κB and JNK activation, suggesting that DGP is a potential prophylactic agent in various neurodegenerative disorders.
Collapse
Affiliation(s)
- Bangrong Cai
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Kyung-Joo Seong
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Sun-Woong Bae
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Changju Chun
- Research Institute of Drug Development, College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Won-Jae Kim
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| | - Ji-Yeon Jung
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
34
|
Thakkar R, Wang R, Wang J, Vadlamudi RK, Brann DW. 17 β-Estradiol Regulates Microglia Activation and Polarization in the Hippocampus Following Global Cerebral Ischemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4248526. [PMID: 29849895 PMCID: PMC5932444 DOI: 10.1155/2018/4248526] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/16/2018] [Accepted: 02/13/2018] [Indexed: 02/08/2023]
Abstract
17β-Estradiol (E2) is a well-known neuroprotective hormone, but its role in regulation of neuroinflammation is less understood. Recently, our lab demonstrated that E2 could regulate the NLRP3 (NOD-like receptor protein 3) inflammasome pathway in the hippocampus following global cerebral ischemia (GCI). Here, we examined the ability of E2 to regulate activation and polarization of microglia phenotype in the hippocampus after global cerebral ischemia (GCI). Our in vivo study in young adult ovariectomized rats showed that exogenous low-dose E2 profoundly suppressed microglia activation and quantitatively shifted microglia from their "activated," amoeboid morphology to a "resting," ramified morphology after GCI. Further studies using M1 "proinflammatory" and M2 "anti-inflammatory" phenotype markers showed that E2 robustly suppressed the "proinflammatory" M1 phenotype, while enhancing the "anti-inflammatory" M2 microglia phenotype in the hippocampus after GCI. These effects of E2 may be mediated directly upon microglia, as E2 suppressed the M1 while enhancing the M2 microglia phenotype in LPS- (lipopolysaccharide-) activated BV2 microglia cells in vitro. E2 also correspondingly suppressed proinflammatory while enhancing anti-inflammatory cytokine gene expression in the LPS-treated BV2 microglia cells. Finally, E2 treatment abolished the LPS-induced neurotoxic effects of BV2 microglia cells upon hippocampal HT-22 neurons. Collectively, our study findings suggest a novel E2-mediated neuroprotective effect via regulation of microglia activation and promotion of the M2 "anti-inflammatory" phenotype in the brain.
Collapse
Affiliation(s)
- Roshni Thakkar
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ruimin Wang
- Department of Neurobiology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, USA
| | - Darrell W. Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
35
|
Slowik A, Lammerding L, Hoffmann S, Beyer C. Brain inflammasomes in stroke and depressive disorders: Regulation by oestrogen. J Neuroendocrinol 2018; 30. [PMID: 28477436 DOI: 10.1111/jne.12482] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 12/28/2022]
Abstract
Neuroinflammation is a devastating pathophysiological process that results in brain damage and neuronal death. Pathogens, cell fragments and cellular dysfunction trigger inflammatory responses. Irrespective of the cause, inflammasomes are key intracellular multiprotein signalling platforms that sense neuropathological conditions. The activation of inflammasomes leads to the auto-proteolytic cleavage of caspase-1, resulting in the proteolysis of the pro-inflammatory cytokines interleukin (IL)1β and IL18 into their bioactive forms. It also initiates pyroptosis, a type of cell death. The two cytokines contribute to the pathogenesis in acute and chronic brain diseases and also play a central role in human aging and psychiatric disorders. Sex steroids, in particular oestrogens, are well-described neuroprotective agents in the central nervous system. Oestrogens improve the functional outcome after ischaemia and traumatic brain injury, reduce neuronal death in Parkinson's and Alzheimer's disease, as well as in amyotrophic lateral sclerosis, attenuate glutamate excitotoxicity and the formation of radical oxygen species, and lessen the spread of oedema after damage. Moreover, oestrogens alleviate menopause-related depressive symptoms and have a positive influence on depressive disorders probably by influencing growth factor production and serotonergic brain circuits. Recent evidence also suggests that inflammasome signalling affects anxiety- and depressive-like behaviour and that oestrogen ameliorates depression-like behaviour through the suppression of inflammasomes. In the present review, we highlight the most recent findings demonstrating that oestrogens selectively suppress the activation of the neuroinflammatory cascade in the brain in acute and chronic brain disease models. Furthermore, we aim to describe putative regulatory signalling pathways involved in the control of inflammasomes. Finally, we consider that psychiatric disorders such as depression also contain an inflammatory component that could be modulated by oestrogen.
Collapse
Affiliation(s)
- A Slowik
- Medical Faculty, Institute of Neuroanatomy, RWTH Aachen, Aachen, Germany
| | - L Lammerding
- Medical Faculty, Institute of Neuroanatomy, RWTH Aachen, Aachen, Germany
| | - S Hoffmann
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen, Germany
- JARA - Translational Brain Medicine, Aachen, Germany
| | - C Beyer
- Medical Faculty, Institute of Neuroanatomy, RWTH Aachen, Aachen, Germany
- JARA - Translational Brain Medicine, Aachen, Germany
| |
Collapse
|
36
|
Zendedel A, Mönnink F, Hassanzadeh G, Zaminy A, Ansar MM, Habib P, Slowik A, Kipp M, Beyer C. Estrogen Attenuates Local Inflammasome Expression and Activation after Spinal Cord Injury. Mol Neurobiol 2017; 55:1364-1375. [PMID: 28127698 DOI: 10.1007/s12035-017-0400-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
Abstract
17-estradiol (E2) is a neuroprotective hormone with a high anti-inflammatory potential in different neurological disorders. The inflammatory response initiated by spinal cord injury (SCI) involves the processing of interleukin-1beta (IL-1b) and IL-18 mediated by caspase-1 which is under the control of an intracellular multiprotein complex called inflammasome. We recently described in a SCI model that between 24 and 72 h post-injury, most of inflammasome components including IL-18, IL-1b, NLRP3, ASC, and caspase-1 are upregulated. In this study, we investigated the influence of E2 treatment after spinal cord contusion on inflammasome regulation. After contusion of T9 spinal segment, 12-week-old male Wistar rats were treated subcutaneously with E2 immediately after injury and every 12 h for the next 3 days. Behavioral scores were significantly improved in E2-treated animals compared to vehicle-treated groups. Functional improvement in E2-treated animals was paralleled by the attenuated expression of certain inflammasome components such as ASC, NLRP1b, and NLRP3 together with IL1b, IL-18, and caspase-1. On the histopathological level, microgliosis and oligodendrocyte injury was ameliorated. These findings support and extend the knowledge of the E2-mediated neuroprotective function during SCI. The control of the inflammasome machinery by E2 might be a missing piece of the puzzle to understand the anti-inflammatory potency of E2.
Collapse
Affiliation(s)
- Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fabian Mönnink
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arash Zaminy
- Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Malek Masoud Ansar
- Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pardes Habib
- Department of Neurology, RWTH Aachen, 52074, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,JARA-Brain, 52074, Aachen, Germany
| |
Collapse
|
37
|
Argente-Arizón P, Guerra-Cantera S, Garcia-Segura LM, Argente J, Chowen JA. Glial cells and energy balance. J Mol Endocrinol 2017; 58:R59-R71. [PMID: 27864453 DOI: 10.1530/jme-16-0182] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
Abstract
The search for new strategies and drugs to abate the current obesity epidemic has led to the intensification of research aimed at understanding the neuroendocrine control of appetite and energy expenditure. This intensified investigation of metabolic control has also included the study of how glial cells participate in this process. Glia, the most abundant cell type in the central nervous system, perform a wide spectrum of functions and are vital for the correct functioning of neurons and neuronal circuits. Current evidence indicates that hypothalamic glia, in particular astrocytes, tanycytes and microglia, are involved in both physiological and pathophysiological mechanisms of appetite and metabolic control, at least in part by regulating the signals reaching metabolic neuronal circuits. Glia transport nutrients, hormones and neurotransmitters; they secrete growth factors, hormones, cytokines and gliotransmitters and are a source of neuroprogenitor cells. These functions are regulated, as glia also respond to numerous hormones and nutrients, with the lack of specific hormonal signaling in hypothalamic astrocytes disrupting metabolic homeostasis. Here, we review some of the more recent advances in the role of glial cells in metabolic control, with a special emphasis on the differences between glial cell responses in males and females.
Collapse
Affiliation(s)
- Pilar Argente-Arizón
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Guerra-Cantera
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jesús Argente
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
38
|
Heitzer M, Kaiser S, Kanagaratnam M, Zendedel A, Hartmann P, Beyer C, Johann S. Administration of 17β-Estradiol Improves Motoneuron Survival and Down-regulates Inflammasome Activation in Male SOD1(G93A) ALS Mice. Mol Neurobiol 2016; 54:8429-8443. [PMID: 27957680 DOI: 10.1007/s12035-016-0322-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease manifested by the progressive loss of upper and lower motoneurons. The pathomechanism of ALS is complex and not yet fully understood. Neuroinflammation is believed to significantly contribute to disease progression. Inflammasome activation was recently shown in the spinal cord of human sporadic ALS patients and in the SOD1(G93A) mouse model for ALS. In the present study, we investigated the neuroprotective and anti-inflammatory effects of 17β-estradiol (E2) treatment in pre-symptomatic and symptomatic male SOD1(G93A) mice. Symptomatic mice with E2 substitution exhibited improved motor performance correlating with an increased survival of motoneurons in the lumbar spinal cord. Expression of NLRP3 inflammasome proteins and levels of activated caspase 1 and mature interleukin 1 beta were significantly reduced in SOD1(G93A) mice supplemented with E2.
Collapse
Affiliation(s)
- Marius Heitzer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sarah Kaiser
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mithila Kanagaratnam
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Philipp Hartmann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,JARA-BRAIN, 52074, Aachen, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
39
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
40
|
Arbo BD, Benetti F, Ribeiro MF. Astrocytes as a target for neuroprotection: Modulation by progesterone and dehydroepiandrosterone. Prog Neurobiol 2016; 144:27-47. [DOI: 10.1016/j.pneurobio.2016.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 01/14/2016] [Accepted: 03/14/2016] [Indexed: 01/19/2023]
|
41
|
Senkyunolide I attenuates oxygen-glucose deprivation/reoxygenation-induced inflammation in microglial cells. Brain Res 2016; 1649:123-131. [PMID: 27524398 DOI: 10.1016/j.brainres.2016.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 01/10/2023]
Abstract
Over-activated microglia during stroke has been documented to aggravate brain damage. Our previous studies showed that senkyunolide I (SEI) exerted anti-inflammatory effects against endotoxin insult in vitro and ameliorative effects on cerebral ischemia/reperfusion (I/R) injury in vivo. Using oxygen-glucose deprivation/reoxygenation (OGD/R) to mimic stroke, we here investigated the anti-inflammatory effect of SEI on microglial cells and explored the underlying mechanisms. OGD for 3h followed by reoxygenation for 12h significantly enhanced the release of pro-inflammatory cytokines and expressions of inflammation-related enzymes in BV-2 cells, which was inhibited by pretreatment with SEI. To elucidate the mechanisms, we studied its effect on upstream signaling pathways. It was found that SEI suppressed the activation of NF-κB pathway induced by OGD/R and the MAPK pathway was shown not to be involved. Furthermore, SEI significantly down-regulated TLR4/MyD88 pathway with specifically improving inducible Hsp70 level through increasing HSF-1/DNA binding activity, and these regulations responsive to SEI were attenuated by transfecting Hsp70 siRNA and HSF-1 decoy ODNs. Additionally, SEI exerted similar influence on Hsp70/TLR4/NF-κB pathway in rat primary microglial cells. The results suggested that SEI had a potent effect against stroke-induced neuroinflammation through suppressing the TLR4/NF-κB pathway by up-regulating Hsp70 dependent on HSF-1.
Collapse
|
42
|
Abstract
Inflammatory activation of microglia is a hallmark of several disorders of the central nervous system. In addition to protecting the brain against inflammatory insults, microglia are neuroprotective and play a significant role in maintaining neuronal connectivity, but the prolongation of an inflammatory status may limit the beneficial functions of these immune cells. The finding that estrogen receptors are present in monocyte-derived cells and that estrogens prevent and control the inflammatory response raise the question of the role that this sex steroid plays in the manifestation and progression of pathologies that have a clear sex difference in prevalence, such as multiple sclerosis, Parkinson's disease, and Alzheimer's disease. The present review aims to provide a critical review of the current literature on the actions of estrogen in microglia and on the involvement of estrogen receptors in the manifestation of selected neurological disorders. This current understanding highlights a research area that should be expanded to identify appropriate replacement therapies to slow the progression of such diseases.
Collapse
Affiliation(s)
- Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
43
|
Kipp M, Hochstrasser T, Schmitz C, Beyer C. Female sex steroids and glia cells: Impact on multiple sclerosis lesion formation and fine tuning of the local neurodegenerative cellular network. Neurosci Biobehav Rev 2016; 67:125-36. [DOI: 10.1016/j.neubiorev.2015.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/30/2015] [Accepted: 11/04/2015] [Indexed: 01/01/2023]
|
44
|
Lei B, Wang H, Jeong S, Hsieh JT, Majeed M, Dawson H, Sheng H, Warner DS, James ML. Progesterone Improves Neurobehavioral Outcome in Models of Intracerebral Hemorrhage. Neuroendocrinology 2016; 103:665-77. [PMID: 26562172 DOI: 10.1159/000442204] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 11/05/2015] [Indexed: 11/19/2022]
Abstract
In models of acute brain injury, progesterone improves recovery through several mechanisms including modulation of neuroinflammation. Secondary injury from neuroinflammation is a potential therapeutic target after intracerebral hemorrhage (ICH). For potential translation of progesterone as a clinical acute ICH therapeutic, the present study sought to define efficacy of exogenous progesterone administration in ICH-relevant experimental paradigms. Young and aged C57BL/6 male, female, and ovariectomized (OVX) mice underwent left intrastriatal collagenase (0.05-0.075 U) or autologous whole blood (35 μl) injection. Progesterone at varying doses (4-16 mg/kg) was administered at 2, 5, 24, 48, and 72 h after injury. Rotarod and Morris water maze latencies were measured on days 1-7 and days 28-31 after injury, respectively. Hematoma volume, brain water content (cerebral edema), complementary immunohistochemistry, multiplex cytokine arrays, and inflammatory proteins were assessed at prespecified time points after injury. Progesterone (4 mg/kg) administration improved rotarod and water maze latencies (p < 0.01), and decreased cerebral edema (p < 0.05), microglial proliferation, and neuronal loss (p < 0.01) in young and aged male, young OVX, and aged female mice. Brain concentration of proinflammatory cytokines and Toll-like receptor-associated proteins were also decreased after progesterone (4 mg/kg) treatment (p < 0.01). Progesterone-treated young female mice showed no detectable effects. Exogenous progesterone improved short- and long-term neurobehavioral recovery and modulated neuroinflammation in male and OVX mice after ICH. Future studies should validate these findings, and address timing and length of administration before translation to clinical trial.
Collapse
Affiliation(s)
- Beilei Lei
- Multidisciplinary Neuroprotection Laboratories, Duke University, Durham, N.C., USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cavaleri F. Review of Amyotrophic Lateral Sclerosis, Parkinson’s and Alzheimer’s diseases helps further define pathology of the novel paradigm for Alzheimer’s with heavy metals as primary disease cause. Med Hypotheses 2015; 85:779-90. [DOI: 10.1016/j.mehy.2015.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/25/2015] [Accepted: 10/11/2015] [Indexed: 01/07/2023]
|
46
|
The progesterone receptor agonist Nestorone holds back proinflammatory mediators and neuropathology in the wobbler mouse model of motoneuron degeneration. Neuroscience 2015; 308:51-63. [DOI: 10.1016/j.neuroscience.2015.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/20/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022]
|
47
|
Slowik A, Beyer C. Inflammasomes are neuroprotective targets for sex steroids. J Steroid Biochem Mol Biol 2015; 153:135-43. [PMID: 25747343 DOI: 10.1016/j.jsbmb.2015.02.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 02/24/2015] [Accepted: 02/27/2015] [Indexed: 01/02/2023]
Abstract
Neuroinflammation in the central nervous system is triggered by toxic stimuli or degenerative events, orchestrates the interplay of brain-intrinsic immune cells and neighboring neural cells, and sequentially allows leukocyte extravasation from the periphery into the brain parenchyma. During the inflammatory cascade, immune-competent cells become activated and secrete a plethora of cytokines and chemokines which form a local inflammatory signaling network important for warding off harmful stimuli to the host but are likewise necessary to preserve damaged brain tissue. Inflammatory responses are initiated by extra- and intra-cellular pathogen and danger-associated receptors. These signals are processed by multi-protein complexes termed inflammasomes which trigger the production of biologically active interleukins-1 and 18 after the cleavage of caspase-1. Estrogens and progesterone are neuroprotective and anti-inflammatory in diverse disease models of the brain in particular under acute inflammatory conditions such as stroke and traumatic brain injury. Both steroids are able to attenuate pro-inflammatory cytokine activity. Recent literature and our own studies provide convincing evidence that the anti-inflammatory potency of these steroids result from a complex interaction with the inflammasome activation and their up-stream regulatory network of miRNAs in brain-intrinsic innate immune cells. This article examines steroid-inflammasome interactions in the brain during brain injury and illuminates the importance of regulation initial upstream events during neuroinflammation. This article is part of a Special Issue entitled 'Steroid Perspectives'.
Collapse
Affiliation(s)
- Alexander Slowik
- Institute of Neuroanatomy, RWTH Aachen University, Medical Faculty, 52074 Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, Medical Faculty, 52074 Aachen, Germany.
| |
Collapse
|
48
|
Khaksari M, Abbasloo E, Dehghan F, Soltani Z, Asadikaram G. The brain cytokine levels are modulated by estrogen following traumatic brain injury: Which estrogen receptor serves as modulator? Int Immunopharmacol 2015; 28:279-87. [PMID: 26112336 DOI: 10.1016/j.intimp.2015.05.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 05/25/2015] [Accepted: 05/28/2015] [Indexed: 12/31/2022]
Abstract
The present study was designed to explore whether administration of estrogen affects brain cytokine levels in TBI. We also sought determine which one of type of classical estrogen receptors (ERs) is involved. Ovariectomized female rats were divided in to eight groups. Estrogen or vehicle was administered following TBI (E2 and oil groups). Antagonist of ER(ICI 182, 780) or vehicle was also administered following TBI (ICI and DMSO groups). The ICI or vehicle was administered either before induction of TBI and administration of estrogen (ICI+E2 and DMSO+E2 groups). TBI was induced by Marmarou's method. In addition to brain water content, the levels of brain proinflammatory and anti-inflammatory cytokines were measured 24 hours post- TBI. Present results demonstrated that, estrogen reduced TBI- induced brain edema. The antiedema effect of estrogen was attenuated by ICI. The brain measures of IL-1β, IL-6 and TNF-α in TBI were also reduced by estrogen. The anti-inflammatory effect of estrogen was attenuated by ICI. The inhibition level of estrogen by ICI was 53.2%, 12.09% and 48.45% for IL-1β, IL-6 and TNF-α, respectively. Estrogen also elevated IL-10 in TBI. ICI inversely controlled the effect of estrogen on IL-10, by 33.84%. This effect was not observed once ICI was used alone. The estrogen administration following TBI probably results in proinflammatory cytokines reduction, and inversely enhancement of anti-inflammatory cytokines. In our study, the neuroprotective effect of estrogen is proposed to be mediated by both ERα and ERα, and accordingly the inhibition of neuroprotective effect of estrogen by ICI.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Abbasloo
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Dehghan
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Asadikaram
- Dept. of Biochemistry, Medical School of Afzalipour, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
49
|
Franco R, Fernández-Suárez D. Alternatively activated microglia and macrophages in the central nervous system. Prog Neurobiol 2015; 131:65-86. [PMID: 26067058 DOI: 10.1016/j.pneurobio.2015.05.003] [Citation(s) in RCA: 495] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 05/22/2015] [Accepted: 05/30/2015] [Indexed: 12/20/2022]
Abstract
Macrophages are important players in the fight against viral, bacterial, fungal and parasitic infections. From a resting state they may undertake two activation pathways, the classical known as M1, or the alternative known as M2. M1 markers are mostly mediators of pro-inflammatory responses whereas M2 markers emerge for resolution and cleanup. Microglia exerts in the central nervous system (CNS) a function similar to that of macrophages in the periphery. Microglia activation and proliferation occurs in almost any single pathology affecting the CNS. Often microglia activation has been considered detrimental and drugs able to stop microglia activation were considered for the treatment of a variety of diseases. Cumulative evidence shows that microglia may undergo the alternative activation pathway, express M2-type markers and contribute to neuroprotection. This review focuses on details about the role of M2 microglia and in the approaches available for its identification. Approaches to drive the M2 phenotype and data on its potential in CNS diseases are also reviewed.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Centro Investigación Biomédica en Red: Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Diana Fernández-Suárez
- Division of Molecular Neurobiology, Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
50
|
Habib P, Beyer C. Regulation of brain microglia by female gonadal steroids. J Steroid Biochem Mol Biol 2015; 146:3-14. [PMID: 24607811 DOI: 10.1016/j.jsbmb.2014.02.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
Microglial cells are the primary mediators of the CNS immune defense system and crucial for shaping inflammatory responses. They represent a highly dynamic cell population which is constantly moving and surveying their environment. Acute brain damage causes a local attraction and activation of this immune cell type which involves neuron-to-glia and glia-to-glia interactions. The prevailing view attributes microglia a "negative" role such as defense and debris elimination. More topical studies also suggest a protective and "positive" regulatory function. Estrogens and progestins exert anti-inflammatory and neuroprotective effects in the CNS in acute and chronic brain diseases. Recent work revealed that microglial cells express subsets of classical and non-classical estrogen and progesterone receptors in a highly dynamic way. In this review article, we would like to stress the importance of microglia for the spreading of neural damage during hypoxia, their susceptibility to functional modulation by sex steroids, the potency of sex hormones to switch microglia from a pro-inflammatory M1 to neuroprotective M2 phenotype, and the regulation of pro- and anti-inflammatory properties including the inflammasome. We will further discuss the possibility that the neuroprotective action of sex steroids in the brain involves an early and direct modulation of local microglia cell function. This article is part of a Special Issue entitled 'Sex steroids and brain disorders'.
Collapse
Affiliation(s)
- Pardes Habib
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|