151
|
Sidibé H, Dubinski A, Vande Velde C. The multi-functional RNA-binding protein G3BP1 and its potential implication in neurodegenerative disease. J Neurochem 2021; 157:944-962. [PMID: 33349931 PMCID: PMC8248322 DOI: 10.1111/jnc.15280] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022]
Abstract
Ras-GTPase-activating protein (GAP)-binding protein 1 (G3BP1) is a multi-functional protein that is best known for its role in the assembly and dynamics of stress granules. Recent studies have highlighted that G3BP1 also has other functions related to RNA metabolism. In the context of disease, G3BP1 has been therapeutically targeted in cancers because its over-expression is correlated with proliferation of cancerous cells and metastasis. However, evidence suggests that G3BP1 is essential for neuronal development and possibly neuronal maintenance. In this review, we will examine the many functions that are carried out by G3BP1 in the context of neurons and speculate how these functions are critical to the progression of neurodegenerative diseases. Additionally, we will highlight the similarities and differences between G3BP1 and the closely related protein G3BP2, which is frequently overlooked. Although G3BP1 and G3BP2 have both been deemed important for stress granule assembly, their roles may differ in other cellular pathways, some of which are specific to the CNS, and presents an opportunity for further exploration.
Collapse
Affiliation(s)
- Hadjara Sidibé
- Department of NeurosciencesUniversité de Montréal, and CHUM Research CenterMontréalQCCanada
| | - Alicia Dubinski
- Department of NeurosciencesUniversité de Montréal, and CHUM Research CenterMontréalQCCanada
| | - Christine Vande Velde
- Department of NeurosciencesUniversité de Montréal, and CHUM Research CenterMontréalQCCanada
| |
Collapse
|
152
|
Activation of microglial G‑protein-coupled receptor 30 protects neurons against excitotoxicity through NF-κB/MAPK pathways. Brain Res Bull 2021; 172:22-30. [PMID: 33848615 DOI: 10.1016/j.brainresbull.2021.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/18/2021] [Accepted: 04/07/2021] [Indexed: 11/21/2022]
Abstract
Neuroexcitotoxicity is a common feature in neuronal damage and neurodegenerative diseases. Our previous studies have confirmed that neuronal and astrocytic G‑protein-coupled receptor 30 (GPR30) play a key role in neuroprotection in vivo and in vitro. Microglia are considered as immune cells in the central nervous system. However, the role of microglial GPR30 in neuroprotection against neuroexcitotoxicity remained unclear. In this study, MTT, Western blot, immunocytochemical staining, phagocytosis assay and wound healing assay were employed to detect the effect of GPR30 in N9 microglial cells after exposure to glutamate. We found that the treatment of GPR30 specific agonist G1 inhibited glutamate-induced proliferation and activation in N9 microglial cells. G1 inhibited M1 polarization, facilitated M2 polarization, and decreased over-phagocytosis but had no effect on migration ability in microglia. The result of neurons and microglia co-culture showed that the activation of microglial GPR30 protected neurons from excitotoxicity through the NF-κB/MAPK signaling pathways. Our findings suggested a key role of microglial GPR30 in excitatory neuronal damage and neurodegenerative diseases.
Collapse
|
153
|
Källstig E, McCabe BD, Schneider BL. The Links between ALS and NF-κB. Int J Mol Sci 2021; 22:3875. [PMID: 33918092 PMCID: PMC8070122 DOI: 10.3390/ijms22083875] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease wherein motor neuron degeneration leads to muscle weakness, progressive paralysis, and death within 3-5 years of diagnosis. Currently, the cause of ALS is unknown but, as with several neurodegenerative diseases, the potential role of neuroinflammation has become an increasingly popular hypothesis in ALS research. Indeed, upregulation of neuroinflammatory factors have been observed in both ALS patients and animal models. One such factor is the inflammatory inducer NF-κB. Besides its connection to inflammation, NF-κB activity can be linked to several genes associated to familial forms of ALS, and many of the environmental risk factors of the disease stimulate NF-κB activation. Collectively, this has led many to hypothesize that NF-κB proteins may play a role in ALS pathogenesis. In this review, we discuss the genetic and environmental connections between NF-κB and ALS, as well as how this pathway may affect different CNS cell types, and finally how this may lead to motor neuron degeneration.
Collapse
Affiliation(s)
| | | | - Bernard L. Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland; (E.K.); (B.D.M.)
| |
Collapse
|
154
|
Hoang NMH, Kim S, Nguyen HD, Kim M, Kim J, Kim BC, Park D, Lee S, Yu BP, Chung HY, Kim MS. Age-Dependent Sensitivity to the Neurotoxic Environmental Metabolite, 1,2-Diacetylbenzene. Biomol Ther (Seoul) 2021; 29:399-409. [PMID: 33820880 PMCID: PMC8255141 DOI: 10.4062/biomolther.2020.208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 01/15/2023] Open
Abstract
1,2-Diacetylbenzene (DAB) is a metabolite of 1,2-diethylbenzene, which is commonly used in the manufacture of plastics and gasoline. We examined the neurotoxic effects of DAB in young and old rats, particularly its effects on hippocampus. Previously, we reported DAB impairs hippocampal neurogenesis but that the underlying mechanism remained unclear. In this study, we evaluate the toxicities exhibited by DAB in the hippocampi of 6-month-old (young) and 20-month-old (old) male SD rats by treating animals intraperitoneally with DAB at 3 mg/kg/day for 1 week. Hippocampal areas were dissected from brains and RNA was extracted and subjected to RNA-seq analysis. RNA results showed animals exhibited age-dependent sensitivity to the neurotoxic effects of DAB. We observed that inflammatory pathways were up-regulated in old rats but that metabolism- and detoxification-related pathways were up-regulated in young rats. This result in old rats, especially upregulation of the TREM1 signaling pathway (an inflammatory response involved in Alzheimer’s disease (AD)) was confirmed by RT-PCR. Our study results provide a better understanding of age-dependent responses to DAB and new insight into the association between DAB and AD.
Collapse
Affiliation(s)
- Ngoc Minh Hong Hoang
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Sungjin Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Minjo Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Jin Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Byoung-Chul Kim
- Systems Toxicology Research Center, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Daeui Park
- Systems Toxicology Research Center, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Sujun Lee
- Department of Pharmacology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| |
Collapse
|
155
|
Chen Y, Tian Z, He L, Liu C, Wang N, Rong L, Liu B. Exosomes derived from miR-26a-modified MSCs promote axonal regeneration via the PTEN/AKT/mTOR pathway following spinal cord injury. Stem Cell Res Ther 2021; 12:224. [PMID: 33820561 PMCID: PMC8022427 DOI: 10.1186/s13287-021-02282-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/11/2021] [Indexed: 12/16/2022] Open
Abstract
Background Exosomes derived from the bone marrow mesenchymal stem cell (MSC) have shown great potential in spinal cord injury (SCI) treatment. This research was designed to investigate the therapeutic effects of miR-26a-modified MSC-derived exosomes (Exos-26a) following SCI. Methods Bioinformatics and data mining were performed to explore the role of miR-26a in SCI. Exosomes were isolated from miR-26a-modified MSC culture medium by ultracentrifugation. A series of experiments, including assessment of Basso, Beattie and Bresnahan scale, histological evaluation, motor-evoked potential recording, diffusion tensor imaging, and western blotting, were performed to determine the therapeutic influence and the underlying molecular mechanisms of Exos-26a in SCI rats. Results Exos-26a was shown to promote axonal regeneration. Furthermore, we found that exosomes derived from miR-26a-modified MSC could improve neurogenesis and attenuate glial scarring through PTEN/AKT/mTOR signaling cascades. Conclusions Exosomes derived from miR-26a-modified MSC could activate the PTEN-AKT-mTOR pathway to promote axonal regeneration and neurogenesis and attenuate glia scarring in SCI and thus present great potential for SCI treatment. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02282-0.
Collapse
Affiliation(s)
- Yuyong Chen
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Zhenming Tian
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Lei He
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Can Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Nangxiang Wang
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Limin Rong
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.
| | - Bin Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
156
|
Adlimoghaddam A, Odero GG, Glazner G, Turner RS, Albensi BC. Nilotinib Improves Bioenergetic Profiling in Brain Astroglia in the 3xTg Mouse Model of Alzheimer's Disease. Aging Dis 2021; 12:441-465. [PMID: 33815876 PMCID: PMC7990369 DOI: 10.14336/ad.2020.0910] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/10/2020] [Indexed: 12/27/2022] Open
Abstract
Current treatments targeting amyloid beta in Alzheimer's disease (AD) have minimal efficacy, which results in a huge unmet medical need worldwide. Accumulating data suggest that brain mitochondrial dysfunction play a critical role in AD pathogenesis. Targeting cellular mechanisms associated with mitochondrial dysfunction in AD create a novel approach for drug development. This study investigated the effects of nilotinib, as a selective tyrosine kinase inhibitor, in astroglia derived from 3xTg-AD mice versus their C57BL/6-controls. Parameters included oxygen consumption rates (OCR), ATP, cytochrome c oxidase (COX), citrate synthase (CS) activity, alterations in oxidative phosphorylation (OXPHOS), nuclear factor kappa B (NF-κB), key regulators of mitochondrial dynamics (mitofusin (Mfn1), dynamin-related protein 1 (Drp1)), and mitochondrial biogenesis (peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α), calcium/calmodulin-dependent protein kinase II (CaMKII), and nuclear factor (erythroid-derived 2)-like 2 (Nrf2)). Nilotinib increased OCR, ATP, COX, Mfn1, and OXPHOS levels in 3xTg astroglia. No significant differences were detected in levels of Drp1 protein and CS activity. Nilotinib enhanced mitochondrial numbers, potentially through a CaMKII-PGC1α-Nrf2 pathway in 3xTg astroglia. Additionally, nilotinib-induced OCR increases were reduced in the presence of the NF-κB inhibitor, Bay11-7082. The data suggest that NF-κB signaling is intimately involved in nilotinib-induced changes in bioenergetics in 3xTg brain astroglia. Nilotinib increased translocation of the NF-κB p50 subunit into the nucleus of 3xTg astroglia that correlates with an increased expression and activation of NF-κB. The current findings support a role for nilotinib in improving mitochondrial function and suggest that astroglia may be a key therapeutic target in treating AD.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada
| | - Gary G Odero
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada
| | - Gordon Glazner
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada.,2Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - R Scott Turner
- 3Department of Neurology, Georgetown University, Washington, DC, USA
| | - Benedict C Albensi
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada.,2Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
157
|
Miao C, Chen H, Li Y, Guo Y, Xu F, Chen Q, Zhang Y, Hu M, Chen G. Curcumin and its analog alleviate diabetes-induced damages by regulating inflammation and oxidative stress in brain of diabetic rats. Diabetol Metab Syndr 2021; 13:21. [PMID: 33602334 PMCID: PMC7891034 DOI: 10.1186/s13098-021-00638-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Diabetic encephalopathy is a severe diabetes complication with cognitive dysfunction and neuropsychiatric disability. The mechanisms underlying diabetic encephalopathy is believed to be relevant with oxidative stress, vascular amylin deposition, immune receptors, inflammation, etc. This study wanted to evaluate the ability of curcumin and its analog A13 to alleviate oxidative stress and inflammation in diabetes-induced damages in brain. METHODS Sixty adult male Sprague-Dawley rats were divided into 5 groups: normal control (NC) group, diabetes mellitus (DM) group, curcumin-treated diabetes mellitus (CUR) group, high dose of A13-treated diabetes mellitus (HA) group, low dose of A13-treated diabetes mellitus (LA) group. Activation of the nuclear factor kappa-B (NF-κB p65) pathway was detected by RT-qPCR, immunohistochemical (IHC) staining and Western blot; oxidative stress was detected by biochemical detection kit; brain tissue sections were stained with hematoxylin-eosin (HE) staining and Myelin staining. RESULTS RT-qPCR, IHC staining and Western blot showed that curcumin and A13 treatment could inhibit the NF-κB p65 pathway. Curcumin and A13 increased the activity of superoxide dismutase and decreased the malondialdehyde level in the brain of diabetic rats. Furthermore, HE staining and Myelin staining demonstrated that the histological lesions of the brain in diabetic rats could be significantly ameliorated by curcumin and A13. CONCLUSION Curcumin analog A13 could alleviate the damages in the brain of diabetes rats by regulating the pathways of inflammation and oxidative stress. A13 may be a new potential therapeutic agent for diabetic encephalopathy.
Collapse
Affiliation(s)
- Chengfeng Miao
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Hanbin Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Yulian Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Ying Guo
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Feifei Xu
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Qi Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Yanyan Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Mengjun Hu
- Department of Pathology, Zhuji People's Hospital, Shaoxing, Zhejiang, PR China.
| | - Guorong Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
158
|
Single-cell brain atlas of Parkinson's disease mouse model. J Genet Genomics 2021; 48:277-288. [PMID: 34052184 DOI: 10.1016/j.jgg.2021.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, leading to the impairment of movement execution. PD pathogenesis has been largely investigated, either limited to bulk transcriptomic levels or at certain cell types, which failed to capture the cellular heterogeneity and intrinsic interplays among distinct cell types. Here, we report the application of single-nucleus RNA-seq on midbrain, striatum, and cerebellum of the α-syn-A53T mouse, a well-established PD mouse model, and matched controls, generating the first single cell transcriptomic atlas for the PD model mouse brain composed of 46,174 individual cells. Additionally, we comprehensively depicte the dysfunctions in PD pathology, covering the elevation of NF-κB activity, the alteration of ion channel components, the perturbation of protein homeostasis network, and the dysregulation of glutamatergic signaling. Notably, we identify a variety of cell types closely associated with PD risk genes. Taken together, our study provides valuable resources to systematically dissect the molecular mechanism of PD pathogenesis at the single-cell resolution, which facilitates the development of novel approaches for diagnosis and therapies against PD.
Collapse
|
159
|
da Costa AEM, Gomes NS, Gadelha Filho CVJ, Linhares MGOES, da Costa RO, Chaves Filho AJM, Cordeiro RC, Vasconcelos GS, da Silva FER, Araujo TDS, Vasconcelos SMM, Lucena DF, Macêdo DS. Sex influences in the preventive effects of peripubertal supplementation with N-3 polyunsaturated fatty acids in mice exposed to the two-hit model of schizophrenia. Eur J Pharmacol 2021; 897:173949. [PMID: 33607108 DOI: 10.1016/j.ejphar.2021.173949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/30/2021] [Accepted: 02/11/2021] [Indexed: 01/05/2023]
Abstract
Schizophrenia is a devastating neurodevelopmental disorder. The animal model based on perinatal immune activation, as first-hit, combined with peripubertal stress, as a second hit, has gained evidence in recent years. Omega-3 polyunsaturated fatty acids (n3-PUFAs) is being a promise for schizophrenia prevention. Nevertheless, the influence of sex in schizophrenia neurobiology and prevention has been neglected. Thus, the present study evaluates the preventive effects of n3-PUFAs in both sexes' mice submitted to the two-hit model and the participation of oxidative changes in this mechanism. The two-hit consisted of polyI:C administration from postnatal days (PNs) 5-7, and unpredictable stress from PNs35-43. n3-PUFAs were administered from PNs30-60. Prepulse inhibition of the startle reflex (PPI), social interaction, and Y-maze tests were conducted between PNs70-72 to evaluate positive-, negative-, and cognitive-like schizophrenia symptoms. We assessed brain oxidative changes in brain areas and plasma. Both sexes' two-hit mice presented deficits in PPI, social interaction, and working memory that were prevented by n3-PUFAs. In two-hit females, n3-PUFAs prevented increments in nitrite levels in the prefrontal cortex (PFC), hippocampus, striatum, and plasma TBARS levels. In two-hit males, n3-PUFAs prevented the increase in TBARS in the PFC, hippocampus, and striatum. Notably, male mice that received only n3-PUFAs without hit exposure presented impairments in working memory and social interaction. These results add further preclinical evidence for n3-PUFAs as an accessible and effective alternative in preventing behavioral and oxidative changes related to schizophrenia but call attention to the need for precaution in this indication due to hit- and sex-sensitive issues.
Collapse
Affiliation(s)
- Ayane Edwiges Moura da Costa
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Carlos Venício Jatai Gadelha Filho
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | | | - Roberta Oliveira da Costa
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Adriano José Maia Chaves Filho
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Rafaela Carneiro Cordeiro
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Germana Silva Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Francisco Eliclécio Rodrigues da Silva
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Tatiane da Silva Araujo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - David Freitas Lucena
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Danielle S Macêdo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
160
|
Yang X, Li F, Liu Y, Li D, Li J. Study on the Correlation Between NF-κB and Central Fatigue. J Mol Neurosci 2021; 71:1975-1986. [PMID: 33586033 PMCID: PMC8500872 DOI: 10.1007/s12031-021-01803-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022]
Abstract
In recent years, the World Health Organization (WHO) has included fatigue as a major risk factor for human life and health. The incidence rate of fatigue is high. In Europe and America, nearly 1/3 of the population is suffering from fatigue. Due to the acceleration of modern people’s life rhythm and the increase of work pressure, more and more attention has been paid to central fatigue. The activation of NF-κB is related to central fatigue, which has been paid little attention by previous studies. At the same time, previous studies have mostly focused on the immune regulation function of NF-κB, while the NF-κB pathway plays an equally important role in regulating nerve function. NF-κB can participate in the occurrence and development of central fatigue by mediating immune inflammatory response, regulating central excitability and inhibitory transmitters, regulating synaptic plasticity and regulating central nervous system (CNS) functional genes. In addition to neuroprotective effects, NF-κB also has nerve damage effects, which is also closely related to the occurrence and development of central fatigue. In this review, we focus on the relationship between NF-κB pathway and central fatigue and further explore the biological mechanism of central fatigue. At the same time, the clinical application and potential of typical NF-κB inhibitors in the treatment of fatigue were analyzed to provide reference for the clinical treatment of central fatigue.
Collapse
Affiliation(s)
- Xingzhe Yang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Feng Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Yan Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Danxi Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
161
|
Diniz WJS, Crouse MS, Cushman RA, McLean KJ, Caton JS, Dahlen CR, Reynolds LP, Ward AK. Cerebrum, liver, and muscle regulatory networks uncover maternal nutrition effects in developmental programming of beef cattle during early pregnancy. Sci Rep 2021; 11:2771. [PMID: 33531552 PMCID: PMC7854659 DOI: 10.1038/s41598-021-82156-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/13/2021] [Indexed: 01/30/2023] Open
Abstract
The molecular basis underlying fetal programming in response to maternal nutrition remains unclear. Herein, we investigated the regulatory relationships between genes in fetal cerebrum, liver, and muscle tissues to shed light on the putative mechanisms that underlie the effects of early maternal nutrient restriction on bovine developmental programming. To this end, cerebrum, liver, and muscle gene expression were measured with RNA-Seq in 14 fetuses collected on day 50 of gestation from dams fed a diet initiated at breeding to either achieve 60% (RES, n = 7) or 100% (CON, n = 7) of energy requirements. To build a tissue-to-tissue gene network, we prioritized tissue-specific genes, transcription factors, and differentially expressed genes. Furthermore, we built condition-specific networks to identify differentially co-expressed or connected genes. Nutrient restriction led to differential tissue regulation between the treatments. Myogenic factors differentially regulated by ZBTB33 and ZNF131 may negatively affect myogenesis. Additionally, nutrient-sensing pathways, such as mTOR and PI3K/Akt, were affected by gene expression changes in response to nutrient restriction. By unveiling the network properties, we identified major regulators driving gene expression. However, further research is still needed to determine the impact of early maternal nutrition and strategic supplementation on pre- and post-natal performance.
Collapse
Affiliation(s)
- Wellison J. S. Diniz
- grid.261055.50000 0001 2293 4611Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND USA
| | - Matthew S. Crouse
- grid.463419.d0000 0001 0946 3608USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE USA
| | - Robert A. Cushman
- grid.463419.d0000 0001 0946 3608USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE USA
| | - Kyle J. McLean
- grid.411461.70000 0001 2315 1184Department of Animal Science, University of Tennessee, Knoxville, TN USA
| | - Joel S. Caton
- grid.261055.50000 0001 2293 4611Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND USA
| | - Carl R. Dahlen
- grid.261055.50000 0001 2293 4611Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND USA
| | - Lawrence P. Reynolds
- grid.261055.50000 0001 2293 4611Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND USA
| | - Alison K. Ward
- grid.261055.50000 0001 2293 4611Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND USA
| |
Collapse
|
162
|
Pajarillo E, Nyarko-Danquah I, Adinew G, Rizor A, Aschner M, Lee E. Neurotoxicity mechanisms of manganese in the central nervous system. ADVANCES IN NEUROTOXICOLOGY 2021; 5:215-238. [PMID: 34263091 DOI: 10.1016/bs.ant.2020.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Getinet Adinew
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Asha Rizor
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
163
|
Regulation of distinct caspase-8 functions in retinal ganglion cells and astroglia in experimental glaucoma. Neurobiol Dis 2021; 150:105258. [PMID: 33434617 DOI: 10.1016/j.nbd.2021.105258] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Retinal ganglion cells (RGCs) expanding from the retina to the brain are primary victims of neurodegeneration in glaucoma, a leading cause of blindness; however, the neighboring astroglia survive the glaucoma-related stress and promote neuroinflammation. In light of diverse functions of caspase-8 in apoptosis, cell survival, and inflammation, this study investigated the importance of caspase-8 in different fates of glaucomatous RGCs and astroglia using two experimental approaches in parallel. In the first approach, cell type-specific responses of RGCs and astroglia to a caspase-8 cleavage-inhibiting pharmacological treatment were studied in rat eyes with or without experimentally induced glaucoma. The second approach utilized an experimental model of glaucoma in mice in which astroglial caspase-8 was conditionally deleted by cre/lox. Findings of these experiments revealed cell type-specific distinct processes that regulate caspase-8 functions in experimental glaucoma, which are involved in inducing the apoptosis of RGCs and promoting the survival and inflammatory responses of astroglia. Deletion of caspase-8 in astroglia protected RGCs against glia-driven inflammatory injury, while the inhibition of caspase-8 cleavage inhibited apoptosis in RGCs themselves. Various caspase-8 functions impacting both RGC apoptosis and astroglia-driven neuroinflammation may suggest the multi-target potential of caspase-8 regulation to provide neuroprotection and immunomodulation in glaucoma.
Collapse
|
164
|
eNOS-dependent S-nitrosylation of the NF-κB subunit p65 has neuroprotective effects. Cell Death Dis 2021; 12:4. [PMID: 33414434 PMCID: PMC7790835 DOI: 10.1038/s41419-020-03338-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 01/29/2023]
Abstract
Cell death by glutamate excitotoxicity, mediated by N-methyl-D-aspartate (NMDA) receptors, negatively impacts brain function, including but not limited to hippocampal neurons. The NF-κB transcription factor (composed mainly of p65/p50 subunits) contributes to neuronal death in excitotoxicity, while its inhibition should improve cell survival. Using the biotin switch method, subcellular fractionation, immunofluorescence, and luciferase reporter assays, we found that NMDA-stimulated NF-κB activity selectively in hippocampal neurons, while endothelial nitric oxide synthase (eNOS), an enzyme expressed in neurons, is involved in the S-nitrosylation of p65 and consequent NF-κB inhibition in cerebrocortical, i.e., resistant neurons. The S-nitro proteomes of cortical and hippocampal neurons revealed that different biological processes are regulated by S-nitrosylation in susceptible and resistant neurons, bringing to light that protein S-nitrosylation is a ubiquitous post-translational modification, able to influence a variety of biological processes including the homeostatic inhibition of the NF-κB transcriptional activity in cortical neurons exposed to NMDA receptor overstimulation.
Collapse
|
165
|
Ibarburu S, Kovacs M, Varela V, Rodríguez-Duarte J, Ingold M, Invernizzi P, Porcal W, Arévalo AP, Perelmuter K, Bollati-Fogolín M, Escande C, López GV, King P, Si Y, Kwon Y, Batthyány C, Barbeito L, Trias E. A Nitroalkene Benzoic Acid Derivative Targets Reactive Microglia and Prolongs Survival in an Inherited Model of ALS via NF-κB Inhibition. Neurotherapeutics 2021; 18:309-325. [PMID: 33118131 PMCID: PMC8116482 DOI: 10.1007/s13311-020-00953-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/28/2022] Open
Abstract
Motor neuron degeneration and neuroinflammation are the most striking pathological features of amyotrophic lateral sclerosis (ALS). ALS currently has no cure and approved drugs have only a modest clinically therapeutic effect in patients. Drugs targeting different deleterious inflammatory pathways in ALS appear as promising therapeutic alternatives. Here, we have assessed the potential therapeutic effect of an electrophilic nitroalkene benzoic acid derivative, (E)-4-(2-nitrovinyl) benzoic acid (BANA), to slow down paralysis progression when administered after overt disease onset in SOD1G93A rats. BANA exerted a significant inhibition of NF-κB activation in NF-κB reporter transgenic mice and microglial cell cultures. Systemic daily oral administration of BANA to SOD1G93A rats after paralysis onset significantly decreased microgliosis and astrocytosis, and significantly reduced the number of NF-κB-p65-positive microglial nuclei surrounding spinal motor neurons. Numerous microglia bearing nuclear NF-κB-p65 were observed in the surrounding of motor neurons in autopsy spinal cords from ALS patients but not in controls, suggesting ALS-associated microglia could be targeted by BANA. In addition, BANA-treated SOD1G93A rats after paralysis onset showed significantly ameliorated spinal motor neuron pathology as well as conserved neuromuscular junction innervation in the skeletal muscle, as compared to controls. Notably, BANA prolonged post-paralysis survival by ~30%, compared to vehicle-treated littermates. These data provide a rationale to therapeutically slow paralysis progression in ALS using small electrophilic compounds such as BANA, through a mechanism involving microglial NF-κB inhibition.
Collapse
Affiliation(s)
- Sofía Ibarburu
- Neurodegeneration Laboratory, Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay
| | - Mariángeles Kovacs
- Neurodegeneration Laboratory, Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay
| | - Valentina Varela
- Neurodegeneration Laboratory, Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay
| | - Jorge Rodríguez-Duarte
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Mariana Ingold
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Paulina Invernizzi
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Williams Porcal
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Ana Paula Arévalo
- Transgenic and Experimental Animal Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Karen Perelmuter
- Cell Biology Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Gloria V López
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Peter King
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, 35294, USA
| | - Ying Si
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, 35294, USA
| | - Yuri Kwon
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Luis Barbeito
- Neurodegeneration Laboratory, Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay.
| | - Emiliano Trias
- Neurodegeneration Laboratory, Institut Pasteur de Montevideo, Mataojo, 2020, Montevideo, Uruguay.
| |
Collapse
|
166
|
Ding ZB, Song LJ, Wang Q, Kumar G, Yan YQ, Ma CG. Astrocytes: a double-edged sword in neurodegenerative diseases. Neural Regen Res 2021; 16:1702-1710. [PMID: 33510058 PMCID: PMC8328766 DOI: 10.4103/1673-5374.306064] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play multifaceted and vital roles in maintaining neurophysiological function of the central nervous system by regulating homeostasis, increasing synaptic plasticity, and sustaining neuroprotective effects. Astrocytes become activated as a result of inflammatory responses during the progression of pathological changes associated with neurodegenerative disorders. Reactive astrocytes (neurotoxic A1 and neuroprotective A2) are triggered during disease progression and pathogenesis due to neuroinflammation and ischemia. However, only a limited body of literature describes morphological and functional changes of astrocytes during the progression of neurodegenerative diseases. The present review investigated the detrimental and beneficial roles of astrocytes in neurodegenerative diseases reported in recent studies, as these cells have promising therapeutic potential and offer new approaches for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhi-Bin Ding
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Yu-Qing Yan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| |
Collapse
|
167
|
The nuclear factor kappa B (NF-κB) signaling pathway is involved in ammonia-induced mitochondrial dysfunction. Mitochondrion 2020; 57:63-75. [PMID: 33378713 DOI: 10.1016/j.mito.2020.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022]
Abstract
Hyperammonemia is very toxic to the brain, leading to inflammation, disruption of brain cellular energy metabolism and cognitive function. However, the underlying mechanism(s) for these impairments is still not fully understood. This study investigated the effects of ammonia in hippocampal astroglia derived from C57BL/6 mice. Parameters measured included oxygen consumption rates (OCR), ATP, cytochrome c oxidase (COX) activity, alterations in oxidative phosphorylation (OXPHOS), nuclear factor kappa B (NF-κB) subunits, key regulators of mitochondrial biogenesis (peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α), calcium/calmodulin-dependent protein kinase II (CaMKII), cAMP-response element binding protein (CREB), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), early growth response (Egr) factor family of proteins, and mitochondrial transcription factor A (TFAM). Ammonia was found to decrease mitochondrial numbers, potentially through a CaMKII-CREB-PGC1α-Nrf2 pathway in astroglia. Ammonia did not alter the levels of Egrs and TFAM in astroglia. Ammonia decreased OCR, ATP, COX, and OXPHOS levels in astroglia. To assess whether energy metabolism is reduced by ammonia through NF-κB associated pathways, astroglia were treated with ammonia alone or with NF-κB inhibitors such as Bay11-7082 or SN50. Mitochondrial OCR levels were reduced in the presence of NF-κB inhibitors; however co-treatment of NF-κB inhibitors and ammonia reversed mitochondrial deficits. Further, ammonia increased translocation of the NF-κB p65 into the nucleus of astroglia that correlates with an increased activity of NF-κB. These findings suggest that the NF-κB signaling pathway is putatively involved in ammonia-induced changes in bioenergetics in astroglia. Such research has critical implications for the treatment of disorders in which brain bioenergetics is compromised.
Collapse
|
168
|
Tong D, Ma Z, Su P, Wang S, Xu Y, Zhang LM, Wu Z, Liu K, Zhao P. Sevoflurane-Induced Neuroapoptosis in Rat Dentate Gyrus Is Activated by Autophagy Through NF-κB Signaling on the Late-Stage Progenitor Granule Cells. Front Cell Neurosci 2020; 14:590577. [PMID: 33384584 PMCID: PMC7769878 DOI: 10.3389/fncel.2020.590577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE The mechanisms by which exposure of the late-stage progenitor cells to the anesthesia sevoflurane alters their differentiation are not known. We seek to query whether the effects of sevoflurane on late-stage progenitor cells might be regulated by apoptosis and/or autophagy. METHODS To address the short-term impact of sevoflurane exposure on granule cell differentiation, we used 5-bromo-2-deoxyuridine (BrdU) to identify the labeled late-stage progenitor granule cells. Male or female rats were exposed to 3% sevoflurane for 4 h when the labeled granule cells were 2 weeks old. Differentiation of the BrdU-labeled granule cells was quantified 4 and 7 days after exposure by double immunofluorescence. The expression of apoptosis and autophagy in hippocampal dentate gyrus (DG) was determined by western blot and immunofluorescence. Western blot for the expression of NF-κB was used to evaluate the mechanism. Morris water maze (MWM) test was performed to detect cognitive function in the rats on postnatal 28-33 days. RESULTS Exposure to sevoflurane decreased the differentiation of the BrdU-labeled late-stage progenitor granule cells, but increased the expression of caspase-3, autophagy, and phosphorylated-P65 in the hippocampus of juvenile rats and resulted in cognitive deficiency. These damaging effects of sevoflurane could be mitigated by inhibitors of autophagy, apoptosis, and NF-κB. The increased apoptosis could be alleviated by pretreatment with the autophagy inhibitor 3-MA, and the increased autophagy and apoptosis could be reduced by pretreatment with NF-κB inhibitor BAY 11-7085. CONCLUSION These findings suggest that a single, prolonged sevoflurane exposure could impair the differentiation of late-stage progenitor granule cells in hippocampal DG and cause cognitive deficits possibly via apoptosis activated by autophagy through NF-κB signaling. Our results do not preclude the possibility that the affected differentiation and functional deficits may be caused by depletion of the progenitors pool.
Collapse
Affiliation(s)
- Dongyi Tong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhongliang Ma
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Su
- Medical Research Center, Shengjing Hospital of China Medical University, Benxi, China
| | - Shuai Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kun Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
169
|
Zika virus exposure affects neuron-glia communication in the hippocampal slices of adult rats. Sci Rep 2020; 10:21604. [PMID: 33303883 PMCID: PMC7729948 DOI: 10.1038/s41598-020-78735-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy was associated with microcephaly in neonates, but clinical and experimental evidence indicate that ZIKV also causes neurological complications in adults. However, the changes in neuron-glial communication, which is essential for brain homeostasis, are still unknown. Here, we report that hippocampal slices from adult rats exposed acutely to ZIKV showed significant cellular alterations regarding to redox homeostasis, inflammatory process, neurotrophic functions and molecular signalling pathways associated with neurons and glial cells. Our findings support the hypothesis that ZIKV is highly neurotropic and its infection readily induces an inflammatory response, characterized by an increased expression and/or release of pro-inflammatory cytokines. We also observed changes in neural parameters, such as adenosine receptor A2a expression, as well as in the release of brain-derived neurotrophic factor and neuron-specific enolase, indicating plasticity synaptic impairment/neuronal damage. In addition, ZIKV induced a glial commitment, with alterations in specific and functional parameters such as aquaporin 4 expression, S100B secretion and glutathione synthesis. ZIKV also induced p21 senescence-associated gene expression, indicating that ZIKV may induce early senescence. Taken together, our results indicate that ZIKV-induced neuroinflammation, involving nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor κB (NFκB) pathways, affects important aspects of neuron-glia communication. Therefore, although ZIKV infection is transient, long-term consequences might be associated with neurological and/or neurodegenerative diseases.
Collapse
|
170
|
An Anthocyanin-Rich Extract Obtained from Portuguese Blueberries Maintains Its Efficacy in Reducing Microglia-Driven Neuroinflammation after Simulated Digestion. Nutrients 2020; 12:nu12123670. [PMID: 33260540 PMCID: PMC7761390 DOI: 10.3390/nu12123670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
Dietary polyphenols are multi-target compounds that have been considered promising candidates in strategies for the mitigation of neurological diseases, acting particularly through reduction of microglia-driven neuroinflammation. In this study, an anthocyanin-rich extract obtained from Portuguese blueberries was subjected to a simulated gastrointestinal digestion; after chemical characterisation, the potential of both non-digested and digested extracts to combat neuroinflammation was evaluated using a microglia N9 cell line. Although the extracts have markedly different chemical composition, both were efficient in reducing the production of either key inflammatory markers or reactive oxygen species and in enhancing reduced glutathione levels in activated cells. Furthermore, this protection was shown to be related to the suppression of nuclear factor kappa B (NF-kB) activation, and to a signal transducer and activator of transcription 1 (STAT1)-independent mechanism. These results demonstrate that the anthocyanin extract, after simulated digestion, maintains its efficacy against neuroinflammation, and can, therefore, assume a relevant role in prevention of neuroinflammation-related neurological disorders.
Collapse
|
171
|
Snow WM, Cadonic C, Cortes-Perez C, Adlimoghaddam A, Roy Chowdhury SK, Thomson E, Anozie A, Bernstein MJ, Gough K, Fernyhough P, Suh M, Albensi BC. Sex-Specific Effects of Chronic Creatine Supplementation on Hippocampal-Mediated Spatial Cognition in the 3xTg Mouse Model of Alzheimer's Disease. Nutrients 2020; 12:nu12113589. [PMID: 33238473 PMCID: PMC7700653 DOI: 10.3390/nu12113589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
The creatine (Cr) energy system has been implicated in Alzheimer’s disease (AD), including reductions in brain phosphoCr and Cr kinase, yet no studies have examined the neurobehavioral effects of Cr supplementation in AD, including the 3xTg mouse model. This studied investigated the effects of Cr supplementation on spatial cognition, plasticity- and disease-related protein levels, and mitochondrial function in the 3xTg hippocampus. Here, 3xTg mice were fed a control or Cr-supplemented (3% Cr (w/w)) diet for 8–9 weeks and tested in the Morris water maze. Mitochondrial oxygen consumption (Seahorse) and protein levels (Western blots) were measured in the hippocampus in subsets of mice. Overall, 3xTg females exhibited impaired memory as compared to males. In females, Cr supplementation decreased escape latency and was associated with increased spatial search strategy use. In males, Cr supplementation decreased the use of spatial search strategies. Pilot data indicated mitochondrial enhancements with Cr supplementation in both sexes. In females, Cr supplementation increased CREB phosphorylation and levels of IκB (NF-κB suppressor), CaMKII, PSD-95, and high-molecular-weight amyloid β (Aβ) species, whereas Aβ trimers were reduced. These data suggest a beneficial preventative effect of Cr supplementation in females and warrant caution against Cr supplementation in males in the AD-like brain.
Collapse
Affiliation(s)
- Wanda M. Snow
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Centre for the Advancement of Teaching and Learning, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: or (W.M.S); (B.C.A.); Tel.: +1-204-235-3942 (B.C.A.)
| | - Chris Cadonic
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Claudia Cortes-Perez
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Aida Adlimoghaddam
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Subir K. Roy Chowdhury
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Research Institute in Oncology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Ella Thomson
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Adama Anozie
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Michael J. Bernstein
- Department of Psychological and Social Sciences, Pennsylvania State University Abington, Abington, PA 19001, USA;
| | - Kathleen Gough
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada;
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Miyoung Suh
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: or (W.M.S); (B.C.A.); Tel.: +1-204-235-3942 (B.C.A.)
| |
Collapse
|
172
|
Peng D, Li J, Deng Y, Zhu X, Zhao L, Zhang Y, Li Z, Ou S, Li S, Jiang Y. Sodium para-aminosalicylic acid inhibits manganese-induced NLRP3 inflammasome-dependent pyroptosis by inhibiting NF-κB pathway activation and oxidative stress. J Neuroinflammation 2020; 17:343. [PMID: 33203418 PMCID: PMC7670624 DOI: 10.1186/s12974-020-02018-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Background The activation of NOD-like receptor protein 3 (NLRP3) inflammasome-dependent pyroptosis has been shown to play a vital role in the pathology of manganese (Mn)-induced neurotoxicity. Sodium para-aminosalicylic acid (PAS-Na) has a positive effect on the treatment of manganism. However, the mechanism is still unclear. We hypothesized that PAS-Na might act through NLRP3. Methods The microglial cell line BV2 and male Sprague-Dawley rats were used to investigate the impacts of PAS-Na on Mn-induced NLRP3 inflammasome-dependent pyroptosis. The related protein of the NF-κB pathway and NLRP3-inflammasome-dependent pyroptosis was detected by western blot. The reactive oxygen species and mitochondrial membrane potential were detected by immunofluorescence staining and flow cytometry. The activation of microglia and the gasdermin D (GSDMD) were detected by immunofluorescence staining. Results Our results showed that Mn treatment induced oxidative stress and activated the NF-κB pathway by increasing the phosphorylation of p65 and IkB-α in BV2 cells and in the basal ganglia of rats. PAS-Na could alleviate Mn-induced oxidative stress damage by inhibiting ROS generation, increasing mitochondrial membrane potential and ATP levels, thereby reducing the phosphorylation of p65 and IkB-α. Besides, Mn treatment could activate the NLRP3 pathway and promote the secretion of IL-18 and IL-1β, mediating pyroptosis in BV2 cells and in the basal ganglia and hippocampus of rats. But an inhibitor of NF-κb (JSH-23) treatment could significantly reduce LDH release, the expression of NLRP3 and Cleaved CASP1 protein and IL-1β and IL-18 mRNA level in BV2 cells. Interestingly, the effect of PAS-Na treatment in Mn-treated BV2 cells is similar to those of JSH-23. Besides, immunofluorescence results showed that PAS-Na reduced the increase number of activated microglia, which stained positively for GSDMD. Conclusion PAS-Na antagonized Mn-induced NLRP3 inflammasome dependent pyroptosis through inhibiting NF-κB pathway activation and oxidative stress. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02018-6.
Collapse
Affiliation(s)
- Dongjie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Junyan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Yue Deng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Xiaojuan Zhu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Lin Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Yuwen Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Zhaocong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Shiyan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China. .,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China. .,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuang-yong Road No.22, Nanning, 530021, Guangxi, China.
| |
Collapse
|
173
|
Henriques JF, Serra D, Dinis TCP, Almeida LM. The Anti-Neuroinflammatory Role of Anthocyanins and Their Metabolites for the Prevention and Treatment of Brain Disorders. Int J Mol Sci 2020; 21:E8653. [PMID: 33212797 PMCID: PMC7696928 DOI: 10.3390/ijms21228653] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Anthocyanins are naturally occurring polyphenols commonly found in fruits and vegetables. Numerous studies have described that anthocyanin-rich foods may play a crucial role in the prevention and treatment of different pathological conditions, which have encouraged their consumption around the world. Anthocyanins exhibit a significant neuroprotective role, mainly due to their well-recognized antioxidant and anti-inflammatory properties. Neuroinflammation is an intricate process relevant in both homeostatic and pathological circumstances. Since the progression of several neurological disorders relies on neuroinflammatory process, targeting brain inflammation has been considered a promising strategy in those conditions. Recent data have shown the anti-neuroinflammatory abilities of many anthocyanins and of their metabolites in the onset and development of several neurological disorders. In this review, it will be discussed the importance and the applicability of these polyphenolic compounds as neuroprotective agents and it will be also scrutinized the molecular mechanisms underlying the modulation of neuroinflammation by these natural compounds in the context of several brain diseases.
Collapse
Affiliation(s)
- Joana F. Henriques
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (J.F.H.); (T.C.P.D.); (L.M.A.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Diana Serra
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (J.F.H.); (T.C.P.D.); (L.M.A.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Teresa C. P. Dinis
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (J.F.H.); (T.C.P.D.); (L.M.A.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Leonor M. Almeida
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (J.F.H.); (T.C.P.D.); (L.M.A.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
174
|
Byun JK, Park M, Lee S, Yun JW, Lee J, Kim JS, Cho SJ, Jeon HJ, Lee IK, Choi YK, Park KG. Inhibition of Glutamine Utilization Synergizes with Immune Checkpoint Inhibitor to Promote Antitumor Immunity. Mol Cell 2020; 80:592-606.e8. [PMID: 33159855 DOI: 10.1016/j.molcel.2020.10.015] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 07/21/2020] [Accepted: 10/11/2020] [Indexed: 02/07/2023]
Abstract
Despite its outstanding clinical success, immune checkpoint blockade remains ineffective in many patients. Accordingly, combination therapy capable of achieving greater antitumor immunity is urgently required. Here, we report that limiting glutamine metabolism in cancer cells bolsters the effectiveness of anti-programmed death ligand-1 (PD-L1) antibody. Inhibition of glutamine utilization increased PD-L1 levels in cancer cells, thereby inactivating co-cultured T cells. Under glutamine-limited conditions, reduced cellular GSH levels caused an upregulation of PD-L1 expression by impairing SERCA activity, which activates the calcium/NF-κB signaling cascade. Consequently, in tumors grown in immunocompetent mice, inhibition of glutamine metabolism decreased the antitumor activity of T cells. In combination with anti-PD-L1, however, glutamine depletion strongly promoted the antitumor efficacy of T cells in vitro and in vivo due to simultaneous increases in Fas/CD95 levels. Our results demonstrate the relevance of cancer glutamine metabolism to antitumor immunity and suggest that co-targeting of glutamine metabolism and PD-L1 represents a promising therapeutic approach.
Collapse
Affiliation(s)
- Jun-Kyu Byun
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea; Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, South Korea
| | - Mihyang Park
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, South Korea
| | - Seunghyeong Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, South Korea
| | - Jae Won Yun
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, South Korea; Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06355, South Korea
| | - Jaebon Lee
- Sungkyunkwan University School of Medicine, Seoul 16419, South Korea
| | - Jae Sun Kim
- Sungkyunkwan University School of Medicine, Seoul 16419, South Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, South Korea
| | - Hui-Jeon Jeon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, South Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea; Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, South Korea; Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, South Korea
| | - Yeon-Kyung Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea; Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, South Korea.
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea; Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, South Korea; Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, South Korea.
| |
Collapse
|
175
|
Robb JL, Morrissey NA, Weightman Potter PG, Smithers HE, Beall C, Ellacott KLJ. Immunometabolic Changes in Glia - A Potential Role in the Pathophysiology of Obesity and Diabetes. Neuroscience 2020; 447:167-181. [PMID: 31765625 PMCID: PMC7567742 DOI: 10.1016/j.neuroscience.2019.10.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Chronic low-grade inflammation is a feature of the pathophysiology of obesity and diabetes in the CNS as well as peripheral tissues. Glial cells are critical mediators of the response to inflammation in the brain. Key features of glia include their metabolic flexibility, sensitivity to changes in the CNS microenvironment, and ability to rapidly adapt their function accordingly. They are specialised cells which cooperate to promote and preserve neuronal health, playing important roles in regulating the activity of neuronal networks across the brain during different life stages. Increasing evidence points to a role of glia, most notably astrocytes and microglia, in the systemic regulation of energy and glucose homeostasis in the course of normal physiological control and during disease. Inflammation is an energetically expensive process that requires adaptive changes in cellular metabolism and, in turn, metabolic intermediates can also have immunomodulatory actions. Such "immunometabolic" changes in peripheral immune cells have been implicated in contributing to disease pathology in obesity and diabetes. This review will discuss the evidence for a role of immunometabolic changes in glial cells in the systemic regulation of energy and glucose homeostasis, and how this changes in the context of obesity and diabetes.
Collapse
Affiliation(s)
- Josephine L Robb
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicole A Morrissey
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Paul G Weightman Potter
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Hannah E Smithers
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
176
|
Xu M, Yan T, Gong G, Wu B, He B, Du Y, Xiao F, Jia Y. Purification, structural characterization, and cognitive improvement activity of a polysaccharides from Schisandra chinensis. Int J Biol Macromol 2020; 163:497-507. [DOI: 10.1016/j.ijbiomac.2020.06.275] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 01/28/2023]
|
177
|
Van den Broek B, Pintelon I, Hamad I, Kessels S, Haidar M, Hellings N, Hendriks JJ, Kleinewietfeld M, Brône B, Timmerman V, Timmermans J, Somers V, Michiels L, Irobi J. Microglial derived extracellular vesicles activate autophagy and mediate multi-target signaling to maintain cellular homeostasis. J Extracell Vesicles 2020; 10:e12022. [PMID: 33708355 PMCID: PMC7890546 DOI: 10.1002/jev2.12022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 12/27/2022] Open
Abstract
Microglia, the immunocompetent cells of the central nervous system (CNS), play an important role in maintaining cellular homeostasis in the CNS. These cells secrete immunomodulatory factors including nanovesicles and participate in the removal of cellular debris by phagocytosis or autophagy. Accumulating evidence indicates that specifically the cellular exchange of small extracellular vesicles (EVs), participates in physiology and disease through intercellular communication. However, the contribution of microglial-derived extracellular vesicles (M-EVs) to the maintenance of microglia homeostasis and how M-EVs could influence the phenotype and gene function of other microglia subtypes is unclear. In addition, knowledge of canonical signalling pathways of inflammation and immunity gene expression patterns in human microglia exposed to M-EVs is limited. Here, we analysed the effects of M-EVs produced in vitro by either tumour necrosis factor alpha (TNFα) activated or non-activated microglia BV2 cells. We showed that M-EVs are internalized by both mouse and human C20 microglia cells and that the uptake of M-EVs in microglia induced autophagic vesicles at various stages of degradation including autophagosomes and autolysosomes. Consistently, stimulation of microglia with M-EVs increased the protein expression of the autophagy marker, microtubule-associated proteins 1A/1B light chain 3B isoform II (LC3B-II), and promoted autophagic flux in live cells. To elucidate the biological activities occurring at the transcriptional level in C20 microglia stimulated with M-EVs, the gene expression profiles, potential upstream regulators, and enrichment pathways were characterized using targeted RNA sequencing. Inflammation and immunity transcriptome gene panel sequencing of both activated and normal microglia stimulated with M-EVs showed involvement of several canonical pathways and reduced expression of key genes involved in neuroinflammation, inflammasome and apoptosis signalling pathways compared to control cells. In this study, we provide the perspective that a beneficial activity of in vitro cell culture produced EVs could be the modulation of autophagy during cellular stress. Therefore, we use a monoculture system to study microglia-microglia crosstalk which is important in the prevention and propagation of inflammation in the brain. We demonstrate that in vitro produced microglial EVs are able to influence multiple biological pathways and promote activation of autophagy in order to maintain microglia survival and homeostasis.
Collapse
Affiliation(s)
| | - Isabel Pintelon
- Laboratory of Cell Biology & HistologyAntwerp Centre for Advanced Microscopy (ACAM)University of AntwerpAntwerpBelgium
| | - Ibrahim Hamad
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
- VIB Laboratory of Translational ImmunomodulationVIB Center for Inflammation ResearchHasseltBelgium
| | - Sofie Kessels
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| | - Mansour Haidar
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| | - Niels Hellings
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| | | | - Markus Kleinewietfeld
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
- VIB Laboratory of Translational ImmunomodulationVIB Center for Inflammation ResearchHasseltBelgium
| | - Bert Brône
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| | - Vincent Timmerman
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born Bunge and University of AntwerpAntwerpBelgium
| | - Jean‐Pierre Timmermans
- Laboratory of Cell Biology & HistologyAntwerp Centre for Advanced Microscopy (ACAM)University of AntwerpAntwerpBelgium
| | - Veerle Somers
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| | - Luc Michiels
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| | - Joy Irobi
- Biomedical Research InstituteUHasseltHasselt UniversityHasseltBelgium
| |
Collapse
|
178
|
Ramli NZ, Yahaya MF, Tooyama I, Damanhuri HA. A Mechanistic Evaluation of Antioxidant Nutraceuticals on Their Potential against Age-Associated Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:E1019. [PMID: 33092139 PMCID: PMC7588884 DOI: 10.3390/antiox9101019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/28/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023] Open
Abstract
Nutraceuticals have been extensively studied worldwide due to its neuroprotective effects in in vivo and in vitro studies, attributed by the antioxidative properties. Alzheimer (AD) and Parkinson disease (PD) are the two main neurodegenerative disorders that are discussed in this review. Both AD and PD share the similar involvement of oxidative stress in their pathophysiology. Nutraceuticals exert their antioxidative effects via direct scavenging of free radicals, prevent damage to biomolecules, indirectly stimulate the endogenous antioxidative enzymes and gene expressions, inhibit activation of pro-oxidant enzymes, and chelate metals. In addition, nutraceuticals can act as modulators of pro-survival, pro-apoptotic, and inflammatory signaling pathways. They have been shown to be effective particularly in preclinical stages, due to their multiple mechanisms of action in attenuating oxidative stress underlying AD and PD. Natural antioxidants from food sources and natural products such as resveratrol, curcumin, green tea polyphenols, and vitamin E are promising therapeutic agents in oxidative stress-mediated neurodegenerative disease as they have fewer adverse effects, more tolerable, cheaper, and sustainable for long term consumption.
Collapse
Affiliation(s)
- Nur Zuliani Ramli
- Department of Biochemistry, Faculty of Medicine, UKM Medical Centre, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
- Department of Biomedical Sciences and Therapeutics, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, UKM Medical Centre, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Ikuo Tooyama
- Molecular Neuroscience Research Centre, Shiga University of Medical Sciences, Seta Tsukinowacho, Otsu 520-2192, Shiga, Japan;
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, UKM Medical Centre, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
179
|
Muraleedharan A, Rotem-Dai N, Strominger I, Anto NP, Isakov N, Monsonego A, Livneh E. Protein kinase C eta is activated in reactive astrocytes of an Alzheimer's disease mouse model: Evidence for its immunoregulatory function in primary astrocytes. Glia 2020; 69:697-714. [PMID: 33068318 DOI: 10.1002/glia.23921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the primary cause of age-related dementia. Pathologically, AD is characterized by synaptic loss, the accumulation of β-amyloid peptides and neurofibrillary tangles, glial activation, and neuroinflammation. Whereas extensive studies focused on neurons and activation of microglia in AD, the role of astrocytes has not been well-characterized. Protein kinase C (PKC) was also implicated in AD; however, its role in astrocyte activation was not elucidated. Using the 5XFAD mouse model of AD, we show that PKC-eta (PKCη), an astrocyte-specific stress-activated and anti-apoptotic kinase, plays a role in reactive astrocytes. We demonstrate that PKCη staining is highly enriched in cortical astrocytes in a disease-dependent manner and in the vicinity of amyloid-β peptides plaques. Moreover, activation of PKCη, as indicated by its increased phosphorylation levels, is exhibited mainly in cortical astrocytes derived from adult 5XFAD mice. PKCη activation was associated with elevated levels of reactive astrocytic markers and upregulation of the pro-inflammatory cytokine interleukin 6 (IL-6) compared to littermate controls. Notably, inhibiting the kinase activity of PKCη in 5XFAD astrocyte cultures markedly increased the levels of secreted IL-6-a phenomenon that was also observed in wild-type astrocytes stimulated by inflammatory cytokines (e.g., TNFα, IL-1). Similar increase in the release of IL-6 was also observed upon inhibition of either the mammalian target of rapamycin (mTOR) or the protein phosphatase 2A (PP2A). Our findings suggest that the mTOR-PKCη-PP2A signaling cascade functions as a negative feedback loop of NF-κB-induced IL-6 release in astrocytes. Thus, we identify PKCη as a regulator of neuroinflammation in AD.
Collapse
Affiliation(s)
- Amitha Muraleedharan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute of Biotechnology in the Negev, Zlotowski Neuroscience Center, and Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noa Rotem-Dai
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Itai Strominger
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute of Biotechnology in the Negev, Zlotowski Neuroscience Center, and Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The National Institute of Biotechnology in the Negev, Zlotowski Neuroscience Center, and Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Etta Livneh
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
180
|
Pustchi SE, Avci NG, Akay YM, Akay M. Astrocytes Decreased the Sensitivity of Glioblastoma Cells to Temozolomide and Bay 11-7082. Int J Mol Sci 2020; 21:E7154. [PMID: 32998285 PMCID: PMC7583902 DOI: 10.3390/ijms21197154] [Citation(s) in RCA: 9] [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: 08/03/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant type of astrocytic tumors. GBM patients have a poor prognosis with a median survival of approximately 15 months despite the "Stupp" Regimen and high tumor recurrence due to the tumor resistance to chemotherapy. In this study, we co-cultured GBM cells with human astrocytes in three-dimensional (3D) poly(ethylene glycol) dimethyl acrylate (PEGDA) microwells to mimic the tumor microenvironment. We treated 3D co- and mono-cultured cells with Temozolomide (TMZ) and the nuclear factor-κB (NF-κB) inhibitor Bay 11-7082 and investigated the combined effect of the drugs. We assessed the expressions of glial fibrillary acidic protein (GFAP) and vimentin that play a role in the tumor malignancy and activation of the astrocytes as well as Notch-1 and survivin that play a role in GBM malignancy after the drug treatment to understand how astrocytes induced GBM drug response. Our results showed that in the co-culture, astrocytes increased GBM survival and resistance after combined drug treatment compared to mono-cultures. These data restated the importance of 3D cell culture to mimic the tumor microenvironment for drug screening.
Collapse
MESH Headings
- Antineoplastic Agents, Alkylating/pharmacology
- Astrocytes/cytology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/genetics
- Coculture Techniques/methods
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Glial Fibrillary Acidic Protein/genetics
- Glial Fibrillary Acidic Protein/metabolism
- Humans
- Models, Biological
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuroglia/pathology
- Nitriles/pharmacology
- Primary Cell Culture
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Signal Transduction
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Sulfones/pharmacology
- Survivin/genetics
- Survivin/metabolism
- Temozolomide/pharmacology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Vimentin/genetics
- Vimentin/metabolism
Collapse
Affiliation(s)
| | | | | | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (S.E.P.); (N.G.A.); (Y.M.A.)
| |
Collapse
|
181
|
Ebrahimpour S, Esmaeili A, Dehghanian F, Beheshti S. Effects of quercetin-conjugated with superparamagnetic iron oxide nanoparticles on learning and memory improvement through targeting microRNAs/NF-κB pathway. Sci Rep 2020; 10:15070. [PMID: 32934245 PMCID: PMC7493930 DOI: 10.1038/s41598-020-71678-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
Quercetin-conjugated superparamagnetic iron oxide nanoparticles (QCSPIONs) have an ameliorative effect on diabetes-induced memory impairment. The current study aimed to compare the effect of quercetin (QC) and QCSPIONs on inflammation-related microRNAs and NF-κB signaling pathways in the hippocampus of diabetic rats. The expression levels of miR-146a, miR-9, NF-κB, and NF-κB-related downstream genes, including TNF-α, BACE1, AβPP, Bax, and Bcl-2 were measured using quantitative real-time PCR. To determine the NF-κB activity, immunohistochemical expression of NF-κB/p65 phosphorylation was employed. Computer simulated docking analysis also performed to find the QC target proteins involved in the NF-κB pathway. Results indicate that diabetes significantly upregulated the expression levels of miR-146a, miR-9, TNF-α, NF-κB, and subsequently AβPP, BACE1, and Bax. Expression analysis shows that QCSPIONs are more effective than pure QC in reducing the expression of miR-9. Interestingly, QCSPIONs reduce the pathological activity of NF-κB and subsequently normalize BACE1, AβPP, and the ratio of Bax/Bcl-2 expression better than pure QC. Comparative docking analyses also show the stronger binding affinity of QC to IKK and BACE1 proteins compared to specific inhibitors of each protein. In conclusion, our study suggests the potent efficacy of QCSPIONs as a promising drug delivery system in memory improvement through targeting the NF-κB pathway.
Collapse
Affiliation(s)
- Shiva Ebrahimpour
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, 81746-73441, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, 81746-73441, Isfahan, Iran.
| | - Fariba Dehghanian
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, 81746-73441, Isfahan, Iran
| | - Siamak Beheshti
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, 81746-73441, Isfahan, Iran
| |
Collapse
|
182
|
Bordeleau M, Lacabanne C, Fernández de Cossío L, Vernoux N, Savage JC, González-Ibáñez F, Tremblay MÈ. Microglial and peripheral immune priming is partially sexually dimorphic in adolescent mouse offspring exposed to maternal high-fat diet. J Neuroinflammation 2020; 17:264. [PMID: 32891154 PMCID: PMC7487673 DOI: 10.1186/s12974-020-01914-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background Maternal nutrition is critical for proper fetal development. While increased nutrient intake is essential during pregnancy, an excessive consumption of certain nutrients, like fat, can lead to long-lasting detrimental consequences on the offspring. Animal work investigating the consequences of maternal high-fat diet (mHFD) revealed in the offspring a maternal immune activation (MIA) phenotype associated with increased inflammatory signals. This inflammation was proposed as one of the mechanisms causing neuronal circuit dysfunction, notably in the hippocampus, by altering the brain-resident macrophages—microglia. However, the understanding of mechanisms linking inflammation and microglial activities to pathological brain development remains limited. We hypothesized that mHFD-induced inflammation could prime microglia by altering their specific gene expression signature, population density, and/or functions. Methods We used an integrative approach combining molecular (i.e., multiplex-ELISA, rt-qPCR) and cellular (i.e., histochemistry, electron microscopy) techniques to investigate the effects of mHFD (saturated and unsaturated fats) vs control diet on inflammatory priming, as well as microglial transcriptomic signature, density, distribution, morphology, and ultrastructure in mice. These analyses were performed on the mothers and/or their adolescent offspring at postnatal day 30. Results Our study revealed that mHFD results in MIA defined by increased circulating levels of interleukin (IL)-6 in the mothers. This phenotype was associated with an exacerbated inflammatory response to peripheral lipopolysaccharide in mHFD-exposed offspring of both sexes. Microglial morphology was also altered, and there were increased microglial interactions with astrocytes in the hippocampus CA1 of mHFD-exposed male offspring, as well as decreased microglia-associated extracellular space pockets in the same region of mHFD-exposed offspring of the two sexes. A decreased mRNA expression of the inflammatory-regulating cytokine Tgfb1 and microglial receptors Tmem119, Trem2, and Cx3cr1 was additionally measured in the hippocampus of mHFD-exposed offspring, especially in males. Conclusions Here, we described how dietary habits during pregnancy and nurturing, particularly the consumption of an enriched fat diet, can influence peripheral immune priming in the offspring. We also found that microglia are affected in terms of gene expression signature, morphology, and interactions with the hippocampal parenchyma, in a partially sexually dimorphic manner, which may contribute to the adverse neurodevelopmental outcomes on the offspring.
Collapse
Affiliation(s)
- Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Chloé Lacabanne
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | | | - Nathalie Vernoux
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Julie C Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Fernando González-Ibáñez
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada. .,Département de médecine moléculaire, Université Laval, Québec, QC, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada. .,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada. .,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
183
|
Yang X, Zeng Q, Barış M, Tezel G. Transgenic inhibition of astroglial NF-κB restrains the neuroinflammatory and neurodegenerative outcomes of experimental mouse glaucoma. J Neuroinflammation 2020; 17:252. [PMID: 32859212 PMCID: PMC7456390 DOI: 10.1186/s12974-020-01930-1] [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: 05/15/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Glia-driven neuroinflammation promotes neuron injury in glaucoma that is a chronic neurodegenerative disease of the optic nerve and a leading cause of irreversible blindness. Although therapeutic modulation of neuroinflammation is increasingly viewed as a logical strategy to avoid inflammatory neurotoxicity in glaucoma, current understanding of the molecular regulation of neuroinflammation is incomplete, and the molecular targets for immunomodulation remains unknown. Growing datasets pointed to nuclear factor-kappaB (NF-κB), a key transcriptional activator of inflammation, which was identified to be most affected in glaucomatous astroglia. Using a cell type-specific experimental approach, this study aimed to determine the value of astroglial NF-κB as a potential treatment target for immunomodulation in experimental mouse glaucoma. METHODS Neuroinflammatory and neurodegenerative outcomes of experimental glaucoma were comparatively analyzed in mice with or without cre/lox-based conditional deletion of astroglial IκKβ, which is the main activating kinase involved in IκB degradation through the canonical pathway of NF-κB activation. Glial responses and the inflammatory status of the retina and optic nerve were analyzed by cell morphology and cytokine profiling, and neuron structure and function were analyzed by counting retinal ganglion cell (RGC) axons and somas and recording pattern electroretinography (PERG) responses. RESULTS Analysis of glial inflammatory responses showed immunomodulatory outcomes of the conditional transgenic deletion of IκKβ in astroglia. Various pro-inflammatory cytokines known to be transcriptional targets for NF-κB exhibited decreased production in IκKβ-deleted astroglia, which included TNF-α that can induce RGC apoptosis and axon degeneration during glaucomatous neurodegeneration. Indeed, transgenic modulation of inflammatory responses by astroglial IκKβ deletion reduced neurodegeneration at different neuronal compartments, including both RGC axons and somas, and protected PERG responses. CONCLUSIONS The findings of this study support a key role for astroglial NF-κB in neuroinflammatory and neurodegenerative outcomes of experimental glaucoma and the potential of this transcriptional regulator pathway as a glial treatment target to provide neuroprotection through immunomodulation. By pointing to a potential treatment strategy targeting the astroglia, these experimental findings are promising for future clinical translation through transgenic applications to improve the treatment of this blinding disease.
Collapse
Affiliation(s)
- Xiangjun Yang
- Department of Ophthalmology, Vagelos College of Physicians and Surgeons, Columbia University, Edward S. Harkness Eye Institute, 635 West 165th Street, New York, NY, 10032, USA
| | - Qun Zeng
- Department of Ophthalmology, Vagelos College of Physicians and Surgeons, Columbia University, Edward S. Harkness Eye Institute, 635 West 165th Street, New York, NY, 10032, USA
| | - Mine Barış
- Department of Ophthalmology, Vagelos College of Physicians and Surgeons, Columbia University, Edward S. Harkness Eye Institute, 635 West 165th Street, New York, NY, 10032, USA
| | - Gülgün Tezel
- Department of Ophthalmology, Vagelos College of Physicians and Surgeons, Columbia University, Edward S. Harkness Eye Institute, 635 West 165th Street, New York, NY, 10032, USA.
| |
Collapse
|
184
|
Stevenson R, Samokhina E, Rossetti I, Morley JW, Buskila Y. Neuromodulation of Glial Function During Neurodegeneration. Front Cell Neurosci 2020; 14:278. [PMID: 32973460 PMCID: PMC7473408 DOI: 10.3389/fncel.2020.00278] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
Glia, a non-excitable cell type once considered merely as the connective tissue between neurons, is nowadays acknowledged for its essential contribution to multiple physiological processes including learning, memory formation, excitability, synaptic plasticity, ion homeostasis, and energy metabolism. Moreover, as glia are key players in the brain immune system and provide structural and nutritional support for neurons, they are intimately involved in multiple neurological disorders. Recent advances have demonstrated that glial cells, specifically microglia and astroglia, are involved in several neurodegenerative diseases including Amyotrophic lateral sclerosis (ALS), Epilepsy, Parkinson's disease (PD), Alzheimer's disease (AD), and frontotemporal dementia (FTD). While there is compelling evidence for glial modulation of synaptic formation and regulation that affect neuronal signal processing and activity, in this manuscript we will review recent findings on neuronal activity that affect glial function, specifically during neurodegenerative disorders. We will discuss the nature of each glial malfunction, its specificity to each disorder, overall contribution to the disease progression and assess its potential as a future therapeutic target.
Collapse
Affiliation(s)
- Rebecca Stevenson
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Evgeniia Samokhina
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Ilaria Rossetti
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - John W. Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- International Centre for Neuromorphic Systems, The MARCS Institute for Brain, Behaviour and Development, Penrith, NSW, Australia
| |
Collapse
|
185
|
ALSUntangled 58: Azathioprine. Amyotroph Lateral Scler Frontotemporal Degener 2020; 22:592-594. [PMID: 32814467 DOI: 10.1080/21678421.2020.1809821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
186
|
Simpson DSA, Oliver PL. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants (Basel) 2020; 9:E743. [PMID: 32823544 PMCID: PMC7463655 DOI: 10.3390/antiox9080743] [Citation(s) in RCA: 440] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders, such as Alzheimer's disease, are a global public health burden with poorly understood aetiology. Neuroinflammation and oxidative stress (OS) are undoubtedly hallmarks of neurodegeneration, contributing to disease progression. Protein aggregation and neuronal damage result in the activation of disease-associated microglia (DAM) via damage-associated molecular patterns (DAMPs). DAM facilitate persistent inflammation and reactive oxygen species (ROS) generation. However, the molecular mechanisms linking DAM activation and OS have not been well-defined; thus targeting these cells for clinical benefit has not been possible. In microglia, ROS are generated primarily by NADPH oxidase 2 (NOX2) and activation of NOX2 in DAM is associated with DAMP signalling, inflammation and amyloid plaque deposition, especially in the cerebrovasculature. Additionally, ROS originating from both NOX and the mitochondria may act as second messengers to propagate immune activation; thus intracellular ROS signalling may underlie excessive inflammation and OS. Targeting key kinases in the inflammatory response could cease inflammation and promote tissue repair. Expression of antioxidant proteins in microglia, such as NADPH dehydrogenase 1 (NQO1), is promoted by transcription factor Nrf2, which functions to control inflammation and limit OS. Lipid droplet accumulating microglia (LDAM) may also represent a double-edged sword in neurodegenerative disease by sequestering peroxidised lipids in non-pathological ageing but becoming dysregulated and pro-inflammatory in disease. We suggest that future studies should focus on targeted manipulation of NOX in the microglia to understand the molecular mechanisms driving inflammatory-related NOX activation. Finally, we discuss recent evidence that therapeutic target identification should be unbiased and founded on relevant pathophysiological assays to facilitate the discovery of translatable antioxidant and anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Dominic S. A. Simpson
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire OX11 0RD, UK;
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Peter L. Oliver
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire OX11 0RD, UK;
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
187
|
Regional, cellular and species difference of two key neuroinflammatory genes implicated in schizophrenia. Brain Behav Immun 2020; 88:826-839. [PMID: 32450195 DOI: 10.1016/j.bbi.2020.05.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
The transcription factor nuclear factor kappa B (NF-κB) regulates the expression of many inflammatory genes that are overexpressed in a subset of people with schizophrenia. Transcriptional reduction in one NF-κB inhibitor, Human Immunodeficiency Virus Enhancer Binding Protein 2 (HIVEP2), is found in the brain of patients, aligning with evidence of NF-κB over-activity. Cellular co-expression of HIVEP2 and cytokine transcripts is a prerequisite for a direct effect of HIVEP2 on pro-inflammatory transcription, and we do not know if changes in HIVEP2 and markers of neuroinflammation are occurring in the same brain cell type. We performed in situ hybridisation on postmortem dorsolateral prefrontal cortex tissue to map and compare the expression of HIVEP2 and Serpin Family A Member 3 (SERPINA3), one of the most consistently increased inflammatory genes in schizophrenia, between schizophrenia patients and controls. We find that HIVEP2 expression is neuronal and is decreased in almost all grey matter cortical layers in schizophrenia patients with neuroinflammation, and that SERPINA3 is increased in the dorsolateral prefrontal cortex grey matter and white matter in the same group of patients. We are the first to map the upregulation of SERPINA3 to astrocytes and to some neurons, and find evidence to suggest that blood vessel-associated astrocytes are the main cellular source of SERPINA3 in the schizophrenia cortex. We show that a lack of HIVEP2 in mice does not cause astrocytic upregulation of Serpina3n but does induce its transcription in neurons. We speculate that HIVEP2 downregulation is not a direct cause of astrocytic pro-inflammatory cytokine synthesis in schizophrenia but may contribute to neuronally-mediated neuroinflammation.
Collapse
|
188
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
189
|
Bidirectional modulation of TNF-α transcription via α- and β-adrenoceptors in cultured astrocytes from rat spinal cord. Biochem Biophys Res Commun 2020; 528:78-84. [DOI: 10.1016/j.bbrc.2020.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/26/2022]
|
190
|
Selvakumar GP, Ahmed ME, Iyer SS, Thangavel R, Kempuraj D, Raikwar SP, Bazley K, Wu K, Khan A, Kukulka K, Bussinger B, Zaheer S, Burton C, James D, Zaheer A. Absence of Glia Maturation Factor Protects from Axonal Injury and Motor Behavioral Impairments after Traumatic Brain Injury. Exp Neurobiol 2020; 29:230-248. [PMID: 32565489 PMCID: PMC7344375 DOI: 10.5607/en20017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) causes disability and death, accelerating the progression towards Alzheimer's disease and Parkinson's disease (PD). TBI causes serious motor and cognitive impairments, as seen in PD that arise during the period of the initial insult. However, this has been understudied relative to TBI induced neuroinflammation, motor and cognitive decline that progress towards PD. Neuronal ubiquitin-C-terminal hydrolase- L1 (UCHL1) is a thiol protease that breaks down ubiquitinated proteins and its level represents the severity of TBI. Previously, we demonstrated the molecular action of glia maturation factor (GMF); a proinflammatory protein in mediating neuroinflammation and neuronal loss. Here, we show that the weight drop method induced TBI neuropathology using behavioral tests, western blotting, and immunofluorescence techniques on sections from wild type (WT) and GMF-deficient (GMF-KO) mice. Results reveal a significant improvement in substantia nigral tyrosine hydroxylase and dopamine transporter expression with motor behavioral performance in GMF-KO mice following TBI. In addition, a significant reduction in neuroinflammation was manifested, as shown by activation of nuclear factor-kB, reduced levels of inducible nitric oxide synthase, and cyclooxygenase- 2 expressions. Likewise, neurotrophins including brain-derived neurotrophic factor and glial-derived neurotrophic factor were significantly improved in GMF-KO mice than WT 72 h post-TBI. Consistently, we found that TBI enhances GFAP and UCHL-1 expression and reduces the number of dopaminergic TH-positive neurons in WT compared to GMF-KO mice 72 h post-TBI. Interestingly, we observed a reduction of THpositive tanycytes in the median eminence of WT than GMF-KO mice. Overall, we found that absence of GMF significantly reversed these neuropathological events and improved behavioral outcome. This study provides evidence that PD-associated pathology progression can be initiated upon induction of TBI.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
191
|
Pellegrino-Coppola D. Regulation of the mitochondrial permeability transition pore and its effects on aging. MICROBIAL CELL (GRAZ, AUSTRIA) 2020; 7:222-233. [PMID: 32904375 PMCID: PMC7453641 DOI: 10.15698/mic2020.09.728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 11/30/2022]
Abstract
Aging is an evolutionarily conserved process and is tightly connected to mitochondria. To uncover the aging molecular mechanisms related to mitochondria, different organisms have been extensively used as model systems. Among these, the budding yeast Saccharomyces cerevisiae has been reported multiple times as a model of choice when studying cellular aging. In particular, yeast provides a quick and trustworthy system to identify shared aging genes and pathway patterns. In this viewpoint on aging and mitochondria, I will focus on the mitochondrial permeability transition pore (mPTP), which has been reported and proposed as a main player in cellular aging. I will make several parallelisms with yeast to highlight how this unicellular organism can be used as a guidance system to understand conserved cellular and molecular events in multicellular organisms such as humans. Overall, a thread connecting the preservation of mitochondrial functionality with the activity of the mPTP emerges in the regulation of cell survival and cell death, which in turn could potentially affect aging and aging-related diseases.
Collapse
|
192
|
Bobermin LD, Roppa RHA, Gonçalves CA, Quincozes-Santos A. Ammonia-Induced Glial-Inflammaging. Mol Neurobiol 2020; 57:3552-3567. [DOI: 10.1007/s12035-020-01985-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
|
193
|
Engelmann C, Riemann M, Carlstedt S, Grimlowski R, Andreas N, Koliesnik I, Meier E, Austerfield P, Haenold R. Identification of undescribed Relb expression domains in the murine brain by new Relb:cre-katushka reporter mice. Dev Dyn 2020; 249:983-997. [PMID: 32145043 DOI: 10.1002/dvdy.170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/07/2020] [Accepted: 02/27/2020] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Noncanonical NF-κB signaling through activation of the transcription factor RelB acts as key regulator of cell lineage determination and differentiation in various tissues including the immune system. To elucidate temporospatial aspects of Relb expression, we generated a BAC transgenic knock-in mouse expressing the fluorescent protein Katushka and the enzyme Cre recombinase under control of the murine Relb promoter (RelbCre-Kat mice). RESULTS Co-expression of Katushka and Relb in fibroblast cultures and tissues of transgenic mice revealed highly specific reporter functions of the transgene. Crossing RelbCre-Kat mice with ROSA26R reporter mice that allow for Cre-mediated consecutive β-galactosidase or YFP synthesis identified various Relb expression domains in perinatal and mature mice. Besides thymus and spleen, highly specific expression patterns were found in different neuronal domains, as well as in other nonimmune organs including skin, skeletal structures and kidney. De novo Relb expression in the mature brain was confirmed in conditional knockout mice with neuro-ectodermal Relb deletion. CONCLUSION Our results demonstrate the usability of RelbCre-Kat reporter mice for the detection of de novo and temporarily restricted Relb expression including cell and lineage tracing of Relb expressing cells. Relb expression during mouse embryogenesis and at adulthood suggests, beyond immunity, important functions of this transcription factor in neurodevelopment and CNS function.
Collapse
Affiliation(s)
| | - Marc Riemann
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Swen Carlstedt
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Jena University Hospital, Institute of Biochemistry II, Center for Sepsis Control and Care, Jena, Germany
| | - Randy Grimlowski
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Nico Andreas
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Jena University Hospital, Institute of Immunology, Jena, Germany
| | - Ievgen Koliesnik
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,School of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Elke Meier
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - Ronny Haenold
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| |
Collapse
|
194
|
Arivazhagan L, Ruiz HH, Wilson R, Manigrasso M, Gugger PF, Fisher EA, Moore KJ, Ramasamy R, Schmidt AM. An Eclectic Cast of Cellular Actors Orchestrates Innate Immune Responses in the Mechanisms Driving Obesity and Metabolic Perturbation. Circ Res 2020; 126:1565-1589. [PMID: 32437306 PMCID: PMC7250004 DOI: 10.1161/circresaha.120.315900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The escalating problem of obesity and its multiple metabolic and cardiovascular complications threatens the health and longevity of humans throughout the world. The cause of obesity and one of its chief complications, insulin resistance, involves the participation of multiple distinct organs and cell types. From the brain to the periphery, cell-intrinsic and intercellular networks converge to stimulate and propagate increases in body mass and adiposity, as well as disturbances of insulin sensitivity. This review focuses on the roles of the cadre of innate immune cells, both those that are resident in metabolic organs and those that are recruited into these organs in response to cues elicited by stressors such as overnutrition and reduced physical activity. Beyond the typical cast of innate immune characters invoked in the mechanisms of metabolic perturbation in these settings, such as neutrophils and monocytes/macrophages, these actors are joined by bone marrow-derived cells, such as eosinophils and mast cells and the intriguing innate lymphoid cells, which are present in the circulation and in metabolic organ depots. Upon high-fat feeding or reduced physical activity, phenotypic modulation of the cast of plastic innate immune cells ensues, leading to the production of mediators that affect inflammation, lipid handling, and metabolic signaling. Furthermore, their consequent interactions with adaptive immune cells, including myriad T-cell and B-cell subsets, compound these complexities. Notably, many of these innate immune cell-elicited signals in overnutrition may be modulated by weight loss, such as that induced by bariatric surgery. Recently, exciting insights into the biology and pathobiology of these cell type-specific niches are being uncovered by state-of-the-art techniques such as single-cell RNA-sequencing. This review considers the evolution of this field of research on innate immunity in obesity and metabolic perturbation, as well as future directions.
Collapse
Affiliation(s)
- Lakshmi Arivazhagan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Henry H. Ruiz
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Robin Wilson
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Michaele Manigrasso
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Paul F. Gugger
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Edward A. Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Langone Medical Center, New York 10016
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, New York 10016
| | - Kathryn J. Moore
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Langone Medical Center, New York 10016
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, New York 10016
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| |
Collapse
|
195
|
Sancho-Balsells A, Brito V, Fernández B, Pardo M, Straccia M, Ginés S, Alberch J, Hernández I, Arranz B, Canals JM, Giralt A. Lack of Helios During Neural Development Induces Adult Schizophrenia-Like Behaviors Associated With Aberrant Levels of the TRIF-Recruiter Protein WDFY1. Front Cell Neurosci 2020; 14:93. [PMID: 32477064 PMCID: PMC7240114 DOI: 10.3389/fncel.2020.00093] [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: 12/31/2019] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
The role of the WDFY1 protein has been studied as a TLR3/4 scaffold/recruiting protein in the immune system and in different oncogenic conditions. However, its function in brain remains poorly understood. We have found that in mice devoid of Helios (He-/- mice), a transcription factor specifically expressed during the development of the immune cells and the central nervous system, there is a permanent and sustained increase of Wdfy1 gene expression in the striatum and hippocampus. Interestingly, we observed that WDFY1 protein levels were also increased in the hippocampus and dorsolateral prefrontal cortex of schizophrenic patients, but not in the hippocampus of Alzheimer's disease patients with an associated psychotic disorder. Accordingly, young He-/- mice displayed several schizophrenic-like behaviors related to dysfunctions in the striatum and hippocampus. These changes were associated with an increase in spine density in medium spiny neurons (MSNs) and with a decrease in the number and size of PSD-95-positive clusters in the stratum radiatum of the CA1. Moreover, these alterations in structural synaptic plasticity were associated with a strong reduction of neuronal NF-κB in the pyramidal layer of the CA1 in He-/- mice. Altogether, our data indicate that alterations involving the molecular axis Helios-WDFY1 in neurons during the development of core brain regions could be relevant for the pathophysiology of neuropsychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Veronica Brito
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Belissa Fernández
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mónica Pardo
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Marco Straccia
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain.,Faculty of Medicine and Health Science, Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Silvia Ginés
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Faculty of Medicine and Health Science, Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Isabel Hernández
- Alzheimer Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades. Barcelona, Spain
| | - Belén Arranz
- Parc Sanitari Sant Joan de Déu, CIBERSAM, Barcelona, Spain
| | - Josep M Canals
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain.,Faculty of Medicine and Health Science, Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Faculty of Medicine and Health Science, Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| |
Collapse
|
196
|
Deng F, Cai L, Zhou B, Zhou Z, Xu G. Whole transcriptome sequencing reveals dexmedetomidine-improves postoperative cognitive dysfunction in rats via modulating lncRNA. 3 Biotech 2020; 10:202. [PMID: 32309111 PMCID: PMC7154046 DOI: 10.1007/s13205-020-02190-9] [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: 01/19/2020] [Accepted: 04/01/2020] [Indexed: 10/24/2022] Open
Abstract
The aim of this study was to explore the underlying mechanism and function of dexmedetomidine (Dex)-regulated long non-coding RNAs (lncRNAs) in improving postoperative cognitive dysfunction (POCD) in rats. The established POCD model, Dex treatment model in rats, Morris water maze testing, and HE staining assays were used to evaluate the efficacy of Dex in POCD treatment in rats. Hippocampus samples of rats from the POCD group and the Dex group were used for lncRNA sequencing. The expression of five differentially expressed lncRNAs (DElncRNAs) was verified by quantitative reverse transcription PCR (qRT-PCR). Competing endogenous RNAs (ceRNA) network was constructed using Cytoscape. The concentration of inflammatory cytokines were measured by ELISA. Microglia proliferation and apoptosis were assessed using CCK-8 assay and flow cytometry, respectively. In the Dex group, the escape latency was shorter, neuron cell injury levels were alleviated, and the expression levels of TNF-α and IL-1β were significantly down-regulated compared with the POCD group. A total of 60 DE lncRNAs were identified, including 16 up- and 44 down-regulated lncRNAs in the Dex group. KEGG pathway analysis revealed that DElncRNAs were significantly enriched in cytokine-cytokine receptor interactions, the p53 signaling pathway, and the NF-kappa B signaling pathway. The qRT-PCR results and ceRNA network suggested that the lncRNA LOC102546895 may play a key role in POCD. LOC102546895 inhibited proliferation while promoting apoptosis in microglial cells and promoted the mRNA and protein expression of the target gene Npas4. Our findings showed that Dex alleviated POCD in rats and regulated lncRNAs expression profile in the hippocampus tissues of rats with POCD. In conclusion, our study outcome proposes that Dex-regulated lncRNA LOC102546895 may play a role in rats with POCD through targeting Npas4.
Collapse
Affiliation(s)
- Fumou Deng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Donghu District, Nanchang, Jiangxi China
| | - Lily Cai
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Donghu District, Nanchang, Jiangxi China
| | - Zhidong Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Donghu District, Nanchang, Jiangxi China
| | - GuoHai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Donghu District, Nanchang, Jiangxi China
| |
Collapse
|
197
|
Welcome MO, Mastorakis NE. Stress-induced blood brain barrier disruption: Molecular mechanisms and signaling pathways. Pharmacol Res 2020; 157:104769. [PMID: 32275963 DOI: 10.1016/j.phrs.2020.104769] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/09/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
Stress is a nonspecific response to a threat or noxious stimuli with resultant damaging consequences. Stress is believed to be an underlying process that can trigger central nervous system disorders such as depression, anxiety, and post-traumatic stress disorder. Though the pathophysiological basis is not completely understood, data have consistently shown a pivotal role of inflammatory mediators and hypothalamo-pituitary-adrenal (HPA) axis activation in stress induced disorders. Indeed emerging experimental evidences indicate a concurrent activation of inflammatory signaling pathways and not only the HPA axis, but also, peripheral and central renin-angiotensin system (RAS). Furthermore, recent experimental data indicate that the HPA and RAS are coupled to the signaling of a range of central neuro-transmitter, -mediator and -peptide molecules that are also regulated, at least in part, by inflammatory signaling cascades and vice versa. More recently, experimental evidences suggest a critical role of stress in disruption of the blood brain barrier (BBB), a neurovascular unit that regulates the movement of substances and blood-borne immune cells into the brain parenchyma, and prevents peripheral injury to the brain substance. However, the mechanisms underlying stress-induced BBB disruption are not exactly known. In this review, we summarize studies conducted on the effects of stress on the BBB and integrate recent data that suggest possible molecular mechanisms and signaling pathways underlying stress-induced BBB disruption. Key molecular targets and pharmacological candidates for treatment of stress and related illnesses are also summarized.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria.
| | | |
Collapse
|
198
|
Sex-specific effects of developmental exposure to polychlorinated biphenyls on neuroimmune and dopaminergic endpoints in adolescent rats. Neurotoxicol Teratol 2020; 79:106880. [PMID: 32259577 DOI: 10.1016/j.ntt.2020.106880] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/22/2022]
Abstract
Exposure to environmental contaminants early in life can have long lasting consequences for physiological function. Polychlorinated biphenyls (PCBs) are a group of ubiquitous contaminants that perturb endocrine signaling and have been associated with altered immune function in children. In this study, we examined the effects of developmental exposure to PCBs on neuroimmune responses to an inflammatory challenge during adolescence. Sprague Dawley rat dams were exposed to a PCB mixture (Aroclor 1242, 1248, 1254, 1:1:1, 20 μg/kg/day) or oil control throughout pregnancy, and adolescent male and female offspring were injected with lipopolysaccharide (LPS, 50 μg/kg, ip) or saline control prior to euthanasia. Gene expression profiling was conducted in the hypothalamus, prefrontal cortex, striatum, and midbrain. In the hypothalamus, PCBs increased expression of genes involved in neuroimmune function, including those within the nuclear factor kappa b (NF-κB) complex, independent of LPS challenge. PCB exposure also increased expression of receptors for dopamine, serotonin, and estrogen in this region. In contrast, in the prefrontal cortex, PCB exposure blunted or induced irregular neuroimmune gene expression responses to LPS challenge. Moreover, neither PCB nor LPS exposure altered expression of neurotransmitter receptors throughout the mesocorticolimbic circuit. Almost all effects were present in males but not females, in agreement with the idea that male neuroimmune cells are more sensitive to perturbation and emphasizing the importance of studying both male and female subjects. Given that altered neuroimmune signaling has been implicated in mental health and substance abuse disorders that often begin during adolescence, these results highlight neuroimmune processes as another mechanism by which early life PCBs can alter brain function later in life.
Collapse
|
199
|
Bellucci A, Bubacco L, Longhena F, Parrella E, Faustini G, Porrini V, Bono F, Missale C, Pizzi M. Nuclear Factor-κB Dysregulation and α-Synuclein Pathology: Critical Interplay in the Pathogenesis of Parkinson's Disease. Front Aging Neurosci 2020; 12:68. [PMID: 32265684 PMCID: PMC7105602 DOI: 10.3389/fnagi.2020.00068] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
The loss of dopaminergic neurons of the nigrostriatal system underlies the onset of the typical motor symptoms of Parkinson's disease (PD). Lewy bodies (LB) and Lewy neurites (LN), proteinaceous inclusions mainly composed of insoluble α-synuclein (α-syn) fibrils are key neuropathological hallmarks of the brain of affected patients. Compelling evidence supports that in the early prodromal phases of PD, synaptic terminal and axonal alterations initiate and drive a retrograde degeneration process culminating with the loss of nigral dopaminergic neurons. This notwithstanding, the molecular triggers remain to be fully elucidated. Although it has been shown that α-syn fibrillary aggregation can induce early synaptic and axonal impairment and cause nigrostriatal degeneration, we still ignore how and why α-syn fibrillation begins. Nuclear factor-κB (NF-κB) transcription factors, key regulators of inflammation and apoptosis, are involved in the brain programming of systemic aging as well as in the pathogenesis of several neurodegenerative diseases. The NF-κB family of factors consists of five different subunits (c-Rel, p65/RelA, p50, RelB, and p52), which combine to form transcriptionally active dimers. Different findings point out a role of RelA in PD. Interestingly, the nuclear content of RelA is abnormally increased in nigral dopamine (DA) neurons and glial cells of PD patients. Inhibition of RelA exert neuroprotection against (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) MPTP and 1-methyl-4-phenylpyridinium (MPP+) toxicity, suggesting that this factor decreases neuronal resilience. Conversely, the c-Rel subunit can exert neuroprotective actions. We recently described that mice deficient for c-Rel develop a PD-like motor and non-motor phenotype characterized by progressive brain α-syn accumulation and early synaptic changes preceding the frank loss of nigrostriatal neurons. This evidence supports that dysregulations in this transcription factors may be involved in the onset of PD. This review highlights observations supporting a possible interplay between NF-κB dysregulation and α-syn pathology in PD, with the aim to disclose novel potential mechanisms involved in the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luigi Bubacco
- Department of Biology, University of Padua, Padua, Italy
| | - Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Edoardo Parrella
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vanessa Porrini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Bono
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Missale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
200
|
Pinitol Prevents Lipopolysaccharide (LPS)-Induced Inflammatory Responses in BV2 Microglia Mediated by TREM2. Neurotox Res 2020; 38:96-104. [DOI: 10.1007/s12640-020-00187-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/12/2020] [Accepted: 02/25/2020] [Indexed: 12/16/2022]
|