1
|
Wood LB, Singer AC. Neurons as Immunomodulators: From Rapid Neural Activity to Prolonged Regulation of Cytokines and Microglia. Annu Rev Biomed Eng 2025; 27:55-72. [PMID: 39805040 DOI: 10.1146/annurev-bioeng-110122-120158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Regulation of the brain's neuroimmune system is central to development, normal function, and disease. Neuronal communication to microglia, the primary immune cells of the brain, is well known to involve purinergic signaling mediated via ATP secretion and the cytokine fractalkine. Recent evidence shows that neurons release multiple cytokines beyond fractalkine, yet these are less studied and poorly understood. In contrast to ATP, cytokines are a class of signaling molecule that are much larger, with longer signaling and farther diffusion. We posit that neuron-expressed cytokines are an essential mechanism of neuron-microglia communication that arises as part of both normal learning and memory and in response to tissue pathology. Thus, neurons are underappreciated immunomodulatory cells that express diverse immunomodulatory signals. While neuronally sourced cytokines have been understudied, new technical advances make this a timely topic. The goal of this review is to define what is known about the cytokines expressed from neurons, how they are regulated, and the effects of these cytokines on microglia. We delineate key knowledge gaps and needs for new tools to define and analyze neuronal roles in immunomodulation. Given that cytokines are central regulators of microglial function, a broad new body of work is required to illuminate functional links between these neuronally expressed cytokines and sustained and transient microglial function.
Collapse
Affiliation(s)
- Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA; ,
| | - Annabelle C Singer
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA; ,
| |
Collapse
|
2
|
Savoca G, Gianfredi A, Bartolini L. The Development of Epilepsy Following CNS Viral Infections: Mechanisms. Curr Neurol Neurosci Rep 2024; 25:2. [PMID: 39549124 DOI: 10.1007/s11910-024-01393-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/18/2024]
Abstract
PURPOSE OF REVIEW This review examines the role of different viral infections in epileptogenesis, with a focus on Herpesviruses such as Human Herpesvirus 6 (HHV-6) and Epstein Barr Virus (EBV), Flaviviruses, Picornaviruses, Human Immunodeficiency Virus (HIV), Influenzavirus and Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2). RECENT FINDINGS A growing literature on animal models, such as the paradigmatic Theiler's murine encephalomyelitis virus (TMEV) model, and clinical investigations in patients with epilepsy have started to elucidate cellular mechanisms implicated in seizure initiation and development of epilepsy following viral infections. A central role of neuroinflammation has emerged, with evidence of activation of the innate and adaptive immunity, dysregulation of microglial and astrocytic activity and production of multiple cytokines and other inflammatory mediators. Several chronic downstream effects result in increased blood-brain barrier permeability, direct neuronal damage, and modifications of ion channels ultimately leading to altered neuronal excitability and seizure generation. Key findings underscore the complex interplay between initial viral infection, neuroinflammation, and later development of epilepsy. Further research is needed to elucidate these mechanisms and develop targeted interventions.
Collapse
Affiliation(s)
- Giulia Savoca
- Neuroscience Department, Meyer Children's Hospital IRCCS, Viale Pieraccini 24, 50139, Florence, Italy
- University of Florence School of Medicine, Florence, Italy
| | - Arianna Gianfredi
- Neuroscience Department, Meyer Children's Hospital IRCCS, Viale Pieraccini 24, 50139, Florence, Italy
- University of Florence School of Medicine, Florence, Italy
| | - Luca Bartolini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Viale Pieraccini 24, 50139, Florence, Italy.
- Department of Neuroscience, Psychology, Pharmacology and Child Health (NEUROFARBA), University of Florence, Florence, Italy.
| |
Collapse
|
3
|
Kaltschmidt B, Czaniera NJ, Schulten W, Kaltschmidt C. NF-κB in Alzheimer's Disease: Friend or Foe? Opposite Functions in Neurons and Glial Cells. Int J Mol Sci 2024; 25:11353. [PMID: 39518906 PMCID: PMC11545113 DOI: 10.3390/ijms252111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a devasting neurodegenerative disease afflicting mainly glutamatergic neurons together with a massive neuroinflammation mediated by the transcription factor NF-κB. A 65%-plus increase in Alzheimer's patients by 2050 might be a major threat to society. Hallmarks of AD are neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and amyloid beta (Aβ) plaques. Here, we review the potential involvement of transcription factor NF-κB by hereditary mutations of the tumor necrosis factor pathway in AD patients. One of the greatest genetic risk factors is APOE4. Recently, it was shown that the APOE4 allele functions as a null allele in human astrocytes not repressing NF-κB anymore. Moreover, NF-κB seems to be involved in the repair of DNA double-strand breaks during healthy learning and memory, a function blunted in AD. NF-κB could be a friend to healthy neurons by repressing apoptosis and necroptosis. But a loss of neuronal NF-κB and activation of glial NF-κB in AD makes it a foe of neuronal survival. Hopeful therapies include TNFR2 receptor bodies relieving the activation of glial NF-κB by TNFα.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| | - Nele Johanne Czaniera
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Wiebke Schulten
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| |
Collapse
|
4
|
Heir R, Abbasi Z, Komal P, Altimimi HF, Franquin M, Moschou D, Chambon J, Stellwagen D. Astrocytes Are the Source of TNF Mediating Homeostatic Synaptic Plasticity. J Neurosci 2024; 44:e2278222024. [PMID: 38395613 PMCID: PMC10993029 DOI: 10.1523/jneurosci.2278-22.2024] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor necrosis factor α (TNF) mediates homeostatic synaptic plasticity (HSP) in response to chronic activity blockade, and prior work has established that it is released from glia. Here we demonstrate that astrocytes are the necessary source of TNF during HSP. Hippocampal cultures from rats of both sexes depleted of microglia still will increase TNF levels following activity deprivation and still express TTX-driven HSP. Slice cultures from mice of either sex with a conditional deletion of TNF from microglia also express HSP, but critically, slice cultures with a conditional deletion of TNF from astrocytes do not. In astrocytes, glutamate signaling is sufficient to reduce NFκB signaling and TNF mRNA levels. Further, chronic TTX treatment increases TNF in an NFκB-dependent manner, although NFκB signaling is dispensable for the neuronal response to TTX-driven HSP. Thus, astrocytes can sense neuronal activity through glutamate spillover and increase TNF production when activity falls, to drive HSP through the production of TNF.
Collapse
Affiliation(s)
- Renu Heir
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Zahra Abbasi
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Pragya Komal
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Haider F Altimimi
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Marie Franquin
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Dionysia Moschou
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Julien Chambon
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| |
Collapse
|
5
|
Akhilesh, Chouhan D, Ummadisetty O, Verma N, Tiwari V. Bergenin ameliorates chemotherapy-induced neuropathic pain in rats by modulating TRPA1/TRPV1/NR2B signalling. Int Immunopharmacol 2023; 125:111100. [PMID: 38149571 DOI: 10.1016/j.intimp.2023.111100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 12/28/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is one of the most prominent and incapacitating complication associated with chemotherapeutic regimens. The exact mechanisms underlying CINP are not fully understood yet, which hampers the development of effective therapeutics. The current study has been designed to investigate the effect of bergenin on CINP and dissect the underlying cellular and molecular mechanisms. Behavioural responsiveness assays were conducted in rats before and after CINP induction and at different time points post-bergenin treatment. We also measured alterations in tight junction proteins, pro-inflammatory cytokines, microglia activity, transient receptor potential (TRP) channels (TRPV1, TRPA1 and TRPM8) and N-methyl-D-aspartate receptor subtype 2 (NR2B) in dorsal root ganglion (DRG) and spinal tissues of neuropathic rats. Bergenin treatment leads to a significant and dose-dependent reduction in evoked and spontaneous ongoing pain without causing central side effects in neuropathic rats. Furthermore, treatment with bergenin and gabapentin did not affect the baseline pain threshold in healthy, non-chemotherapy-treated rats, as evaluated through tail-flick and tail-clip assays. Chemotherapy administration leads to a significant activation of TRP channels, concurrent with microglial activation, disruption of spinal cord tight junction proteins, and subsequent infiltration of pro-inflammatory cytokines, as well as NR2B activation. Notably, bergenin treatment effectively reversed all of these alterations, with the exception of TRPM8, in both the DRG and spinal cord of neuropathic rats. Findings from the present study suggests that bergenin mitigates neuropathic pain by modulating the TRPA1/TRPV1/NR2B signalling and presents a promising therapeutic avenue for the treatment of chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Nivedita Verma
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
6
|
Kaltschmidt B, Helweg LP, Greiner JFW, Kaltschmidt C. NF-κB in neurodegenerative diseases: Recent evidence from human genetics. Front Mol Neurosci 2022; 15:954541. [PMID: 35983068 PMCID: PMC9380593 DOI: 10.3389/fnmol.2022.954541] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The transcription factor NF-κB is commonly known to drive inflammation and cancer progression, but is also a crucial regulator of a broad range of cellular processes within the mammalian nervous system. In the present review, we provide an overview on the role of NF-κB in the nervous system particularly including its constitutive activity within cortical and hippocampal regions, neuroprotection as well as learning and memory. Our discussion further emphasizes the increasing role of human genetics in neurodegenerative disorders, namely, germline mutations leading to defects in NF-κB-signaling. In particular, we propose that loss of function mutations upstream of NF-κB such as ADAM17, SHARPIN, HOIL, or OTULIN affect NF-κB-activity in Alzheimer’s disease (AD) patients, in turn driving anatomical defects such as shrinkage of entorhinal cortex and the limbic system in early AD. Similarly, E3 type ubiquitin ligase PARKIN is positively involved in NF-κB signaling. PARKIN loss of function mutations are most frequently observed in Parkinson’s disease patients. In contrast to AD, relying on germline mutations of week alleles and a disease development over decades, somatic mutations affecting NF-κB activation are commonly observed in cells derived from glioblastoma multiforme (GBM), the most common malignant primary brain tumor. Here, our present review particularly sheds light on the mutual exclusion of either the deletion of NFKBIA or amplification of epidermal growth factor receptor (EGFR) in GBM, both resulting in constitutive NF-κB-activity driving tumorigenesis. We also discuss emerging roles of long non-coding RNAs such as HOTAIR in suppressing phosphorylation of IκBα in the context of GBM. In summary, the recent progress in the genetic analysis of patients, particularly those suffering from AD, harbors the potential to open up new vistas for research and therapy based on TNFα/NF-κB pathway and neuroprotection.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Department of Molecular Neurobiology, Bielefeld University, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
- *Correspondence: Barbara Kaltschmidt,
| | - Laureen P. Helweg
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
| | - Johannes F. W. Greiner
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), Bielefeld, Germany
- Department of Cell Biology, Biological Faculty, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
7
|
Sriram S, Mehkri Y, Quintin S, Lucke-Wold B. Shared pathophysiology: Understanding stroke and Alzheimer's disease. Clin Neurol Neurosurg 2022; 218:107306. [PMID: 35636382 DOI: 10.1016/j.clineuro.2022.107306] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/19/2022] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease and stroke share several known vascular risk factors. The pathophysiology and whether one predisposes to the other is a topic of ongoing investigation. In this critical review, we highlight what is known about each pathway and the shared potential mechanisms. We offer insight into topics that warrant further investigation. We address topics of both neurodegeneration and secondary cascades. Furthermore, the concept of targeting secondary mechanisms early might be a viable treatment option for ongoing preventative measures. The review is intended to serve as a catalyst for further scientific inquiry into this important topic.
Collapse
Affiliation(s)
- Sai Sriram
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | - Yusuf Mehkri
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | - Stephan Quintin
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | | |
Collapse
|
8
|
Löscher W, Howe CL. Molecular Mechanisms in the Genesis of Seizures and Epilepsy Associated With Viral Infection. Front Mol Neurosci 2022; 15:870868. [PMID: 35615063 PMCID: PMC9125338 DOI: 10.3389/fnmol.2022.870868] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 12/16/2022] Open
Abstract
Seizures are a common presenting symptom during viral infections of the central nervous system (CNS) and can occur during the initial phase of infection ("early" or acute symptomatic seizures), after recovery ("late" or spontaneous seizures, indicating the development of acquired epilepsy), or both. The development of acute and delayed seizures may have shared as well as unique pathogenic mechanisms and prognostic implications. Based on an extensive review of the literature, we present an overview of viruses that are associated with early and late seizures in humans. We then describe potential pathophysiologic mechanisms underlying ictogenesis and epileptogenesis, including routes of neuroinvasion, viral control and clearance, systemic inflammation, alterations of the blood-brain barrier, neuroinflammation, and inflammation-induced molecular reorganization of synapses and neural circuits. We provide clinical and animal model findings to highlight commonalities and differences in these processes across various neurotropic or neuropathogenic viruses, including herpesviruses, SARS-CoV-2, flaviviruses, and picornaviruses. In addition, we extensively review the literature regarding Theiler's murine encephalomyelitis virus (TMEV). This picornavirus, although not pathogenic for humans, is possibly the best-characterized model for understanding the molecular mechanisms that drive seizures, epilepsy, and hippocampal damage during viral infection. An enhanced understanding of these mechanisms derived from the TMEV model may lead to novel therapeutic interventions that interfere with ictogenesis and epileptogenesis, even within non-infectious contexts.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Charles L. Howe
- Division of Experimental Neurology, Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
9
|
Khan-Mohammadi-Khorrami MK, Asle-Rousta M, Rahnema M, Amini R. Neuroprotective effect of alpha-pinene is mediated by suppression of the TNF-α/NF-κB pathway in Alzheimer's disease rat model. J Biochem Mol Toxicol 2022; 36:e23006. [PMID: 35174932 DOI: 10.1002/jbt.23006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/19/2021] [Accepted: 01/19/2022] [Indexed: 01/04/2023]
Abstract
Monoterpene alpha-pinene possesses antioxidant, cardioprotective, and neuroprotective properties. We evaluated the effect of alpha-pinene on oxidative/nitrosative stress, neuroinflammation, and molecular and behavioral changes induced by beta-amyloid (Aβ)1-42 in rats and investigated the possible mechanisms of these outcomes. Male Wistar rats received alpha-pinene (50 mg/kg intraperitoneally) for 14 consecutive days after intrahippocampal injection of Aβ1-42 . Alpha-pinene decreased malondialdehyde and nitric oxide levels, increased glutathione content, and enhanced catalase activity in Aβ-injected rats. Also, messenger RNA expression of tumor necrosis factor-α, interleukin-1β, interleukin-6, nuclear factor κB, and N-methyl- d-aspartate receptor subunits 2A and 2B reduced in the hippocampus of these animals. Besides this, alpha-pinene repressed the Aβ1-42 -induced reduction of nicotinic acetylcholine receptor α7 subunit and brain-derived neurotrophic factor expression. Treatment with alpha-pinene caused Aβ-receiving rats to spend more time in the target quadrant in the Morris water maze test and led to an increase in percentages of open arm entrance and time spent in the open arm in the elevated plus-maze test. We concluded that alpha-pinene strengthens the antioxidant system and prevents neuroinflammation in the hippocampus of rats receiving Aβ. It improves spatial learning and memory and reduces anxiety-like behavior in these animals. Consequently, alpha-pinene alleviates Aβ-induced oxidative/nitrosative stress, neuroinflammation, and behavioral deficits. It is probably a suitable candidate for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Mehdi Rahnema
- Department of Physiology, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Rahim Amini
- Department of Biology, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| |
Collapse
|
10
|
Uniyal A, Akhilesh, Singh Rathore A, Kumari Keshri P, Pratap Singh S, Singh S, Tiwari V. Inhibition of pan-Aurora kinase attenuates evoked and ongoing pain in nerve injured rats via regulating KIF17-NR2B mediated signaling. Int Immunopharmacol 2022; 106:108622. [PMID: 35183034 DOI: 10.1016/j.intimp.2022.108622] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
Kinesins (KIF's) are the motor proteins which are recently reported to be involved in the trafficking of nociceptors leading to chronic pain. Aurora kinases are known to be involved in the regulation of KIF proteins which are associated with the activation of N-methyl-D-aspartate (NMDA) receptors. Here, we investigated the effect of tozasertib, a pan-Aurora kinase inhibitor, on nerve injury-induced evoked and chronic ongoing pain in rats and the involvement of kinesin family member 17 (KIF17) and NMDA receptor subtype 2B (NR2B) crosstalk in the same. Rats with chronic constriction injury showed a significantly decreased pain threshold in a battery of pain behavioural assays. We found that tozasertib [10, 20, and 40 mg/kg intraperitoneally (i.p.)] treatment showed a significant and dose-dependent inhibition of both evoked and chronic ongoing pain in rats with nerve injury. Tozasertib (40 mg/kg i.p.) and gabapentin (30 mg/kg i.p.) treatment significantly inhibits spontaneous ongoing pain in nerve injured rats but did not produce any place preference behaviour in healthy naïve rats pointing towards their non-addictive analgesic potential. Moreover, tozasertib (10, 20, and 40 mg/kg i.p.) and gabapentin (30 mg/kg i.p.) treatment did not altered the normal pain threshold in healthy naïve rats and didn't produce central nervous system associated side effects as well. Western blotting and reverse transcription polymerase chain reaction studies suggested enhanced expressions of NR2B and KIF-17 along with increased nuclear factor kappa β (NFkβ), tumour necrosis factor-α (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6) levels in dorsal root ganglion (DRG) and spinal cord of nerve injured rats which was significantly attenuated on treatment with different does of Tozasertib. Findings from the current study suggests that inhibition of pan-Aurora kinase decreased KIF-17 mediated NR2B activation which further leads to significant inhibition of evoked and chronic ongoing pain in nerve-injured rats.
Collapse
Affiliation(s)
- Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Aaina Singh Rathore
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Sanjay Singh
- Baba Saheb Bhim Rao Ambedkar Central University (BBAU), Lucknow 226025, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
11
|
Sanders OD, Rajagopal L, Rajagopal JA. Does oxidatively damaged DNA drive amyloid-β generation in Alzheimer's disease? A hypothesis. J Neurogenet 2021; 35:351-357. [PMID: 34282704 DOI: 10.1080/01677063.2021.1954641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In Alzheimer's disease (AD), amyloid-β (Aβ) generation and upstream β-secretase 1 (BACE1) expression appear to be driven by oxidative stress via c-Jun N-terminal kinase (JNK), p38, and Interferon-Induced, Double-Stranded RNA-Activated Protein Kinase (PKR). In addition, inflammatory molecules, including lipopolysaccharide (LPS), induce genes central to Aβ genesis, such as BACE1, via nuclear factor-κB (NFκB). However, additional triggers of Aβ generation remain poorly understood and might represent novel opportunities for therapeutic intervention. Based on mechanistic studies and elevated ectopic oxidatively damaged DNA (oxoDNA) levels in preclinical AD, mild cognitive impairment, and AD patients, we hypothesize oxoDNA contributes to β-amyloidosis starting from the earliest stages of AD through multiple pathways. OxoDNA induces mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), thereby sensitizing the brain to oxidative stress-induced JNK activation and BACE1 transcription. It also induces myeloid differentiation primary response 88 (MyD88) and activates protein kinase CK2, thereby increasing NFκB activation and BACE1 induction. OxoDNA increases oxidative stress via nuclear factor erythroid 2-related factor 2 (Nrf2) ectopic localization, likely augmenting JNK-mediated BACE1 induction. OxoDNA likely also promotes β-amyloidosis via absent in melanoma 2 (AIM2) induction. Falsifiable predictions of this hypothesis include that deoxyribonuclease treatment should decrease Aβ and possibly slow cognitive decline in AD patients. While formal testing of this hypothesis remains to be performed, a case report has found deoxyribonuclease I treatment improved a severely demented AD patient's Mini-Mental Status Exam score from 3 to 18 at 2 months. There is preliminary preclinical and clinical evidence suggesting that ectopic oxidatively damaged DNA may act as an inflammatory damage-associated molecular pattern contributing to Aβ generation in AD, and deoxyribonuclease I should be formally evaluated to test whether it can decrease Aβ levels and slow cognitive decline in AD patients.
Collapse
|
12
|
Nanoparticle-based fluorescence probe for detection of NF-κB transcription factor in single cell via steric hindrance. Mikrochim Acta 2021; 188:226. [PMID: 34106343 DOI: 10.1007/s00604-021-04878-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
A novel nanoparticle-based fluorescence probe was developed for NF-κB transcription factor detection and in situ imaging via steric hindrance. The probe contains gold nanoparticles (AuNPs) to quench fluorescence, and nucleic acids immobilized on the surface of AuNPs to output fluorescence. In the basal state, Cy5 labeled DNA1 folds its long chain into a hairpin structure and quenches fluorescence by forcing the Cy5 fluorophore close to the surface of AuNPs. After the probe enters the cell, the NF-κB transcription factor can bind to the κB site in the DNA duplex of the nucleic acids. The steric hindrance caused by NF-κB leads to the extension of the long chain of DNA1 and the removal of the Cy5 fluorophore from the surface of AuNPs, thereby restoring the fluorescence of the probe. By measuring NF-κB in cell lysis in vitro, the probe obtains a detection limit of 0.38 nM and the linear range from 0.5 to 16 nM. Repeated measurements showed the recovery in the cell nuclear extract was between 93.38 and 109.32%, with relative standard deviation less than 5%. By monitoring the sub-localization of the Cy5 fluorophore in single cell, the probe system can effectively distinguish active NF-κB (nucleus) and inactive NF-κB (cytoplasm) through in situ imaging. The well-designed probe will make up for the shortcomings of the existing technology, and reveal the regulatory role of transcription factors in many disease processes.
Collapse
|
13
|
Trombetta-Lima M, Assis-Ribas T, Cintra RC, Campeiro JD, Guerreiro JR, Winnischofer SMB, Nascimento ICC, Ulrich H, Hayashi MAF, Sogayar MC. Impact of Reck expression and promoter activity in neuronal in vitro differentiation. Mol Biol Rep 2021; 48:1985-1994. [PMID: 33619662 DOI: 10.1007/s11033-021-06175-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
Reck (REversion-inducing Cysteine-rich protein with Kazal motifs) tumor suppressor gene encodes a multifunctional glycoprotein which inhibits the activity of several matrix metalloproteinases (MMPs), and has the ability to modulate the Notch and canonical Wnt pathways. Reck-deficient neuro-progenitor cells undergo precocious differentiation; however, modulation of Reck expression during progression of the neuronal differentiation process is yet to be characterized. In the present study, we demonstrate that Reck expression levels are increased during in vitro neuronal differentiation of PC12 pheochromocytoma cells and P19 murine teratocarcinoma cells and characterize mouse Reck promoter activity during this process. Increased Reck promoter activity was found upon induction of differentiation in PC12 cells, in accordance with its increased mRNA expression levels in mouse in vitro models. Interestingly, Reck overexpression, prior to the beginning of the differentiation protocol, led to diminished efficiency of the neuronal differentiation process. Taken together, our findings suggest that increased Reck expression at early stages of differentiation diminishes the number of neuron-like cells, which are positive for the beta-3 tubulin marker. Our data highlight the importance of Reck expression evaluation to optimize in vitro neuronal differentiation protocols.
Collapse
Affiliation(s)
- Marina Trombetta-Lima
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo (USP), Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil
| | - Thais Assis-Ribas
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo (USP), Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil
| | - Ricardo C Cintra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Joana D Campeiro
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Rua 3 de Maio 100, Ed INFAR, 3º andar, São Paulo, SP, 04044-020, Brazil
| | - Juliano R Guerreiro
- Faculdade de Farmácia, Universidade Paulista (UNIP), São Paulo, SP, 05347-020, Brazil
| | - Sheila M B Winnischofer
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná (UFPR), Curitiba, PR, 81531-990, Brazil
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba, PR, 81531-990, Brazil
| | - Isis C C Nascimento
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Mirian A F Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Rua 3 de Maio 100, Ed INFAR, 3º andar, São Paulo, SP, 04044-020, Brazil.
| | - Mari C Sogayar
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo (USP), Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil.
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
14
|
Lee-Rivera I, López E, Alvarez-Arce A, López-Colomé AM. The PKC-ζ pseudosubstrate peptide induces glutamate release from retinal pigment epithelium cells through kinase- independent activation of Best1. Life Sci 2020; 265:118860. [PMID: 33301813 DOI: 10.1016/j.lfs.2020.118860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/19/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
AIMS The retinal pigment epithelium (RPE) is a highly specialized cell monolayer, that plays a key role in the maintenance of photoreceptor function and the blood-retina barrier (BRB). In this study, we found that a myristoylated pseudosubstrate of PKC-ζ (PKCζ PS), considered as a PKC-ζ inhibitor, plays a distinct role in RPE. MAIN METHODS We demonstrated that PKCζ PS stimulates the release of Glutamate (Glu) using in vitro3H-Glutamate release experiments. By western blot, kinase assays, and Fluoresence Ca+2 Concentration Measurements, we determined the cellular mechanisms involved in such release. KEY FINDINGS Surprisingly, PKCζ PS has no effect on either phosphorylation of T560, essential for catalytic activity, nor it has an effect on kinase activity. It induces the dose-dependent release of Glu by increasing intracellular Ca+2 levels. Interestingly, this release was not observed upon stimulation by other non-competitive PKC-ζ inhibitors. We here demonstrated that the PKCζ PS stimulates the release of Glutamate from RPE by activating the Ca2+-dependent Cl channel Bestrophin 1 (Best1). SIGNIFICANCE These results question PKCζ PS specificity as an inhibitor of this enzyme. Furthermore, the present results underline the relevance of clarifying the molecular mechanisms involved in glutamate release from the retina under conditions derived from excitotoxic stimuli.
Collapse
Affiliation(s)
- Irene Lee-Rivera
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, UNAM, Apartado Postal 70-253, Ciudad Universitaria, México City, CdMx, Mexico
| | - Edith López
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, UNAM, Apartado Postal 70-253, Ciudad Universitaria, México City, CdMx, Mexico
| | - Alejandro Alvarez-Arce
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, UNAM, Apartado Postal 70-253, Ciudad Universitaria, México City, CdMx, Mexico
| | - Ana María López-Colomé
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, UNAM, Apartado Postal 70-253, Ciudad Universitaria, México City, CdMx, Mexico.
| |
Collapse
|
15
|
Velásquez E, Martins-de-Souza D, Velásquez I, Carneiro GRA, Schmitt A, Falkai P, Domont GB, Nogueira FCS. Quantitative Subcellular Proteomics of the Orbitofrontal Cortex of Schizophrenia Patients. J Proteome Res 2019; 18:4240-4253. [PMID: 31581776 DOI: 10.1021/acs.jproteome.9b00398] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a chronic disease characterized by the impairment of mental functions with a marked social dysfunction. A quantitative proteomic approach using iTRAQ labeling and SRM, applied to the characterization of mitochondria (MIT), crude nuclear fraction (NUC), and cytoplasm (CYT), can allow the observation of dynamic changes in cell compartments providing valuable insights concerning schizophrenia physiopathology. Mass spectrometry analyses of the orbitofrontal cortex from 12 schizophrenia patients and 8 healthy controls identified 655 protein groups in the MIT fraction, 1500 in NUC, and 1591 in CYT. We found 166 groups of proteins dysregulated among all enriched cellular fractions. Through the quantitative proteomic analysis, we detect as the main biological pathways those related to calcium and glutamate imbalance, cell signaling disruption of CREB activation, axon guidance, and proteins involved in the activation of NF-kB signaling along with the increase of complement protein C3. Based on our data analysis, we suggest the activation of NF-kB as a possible pathway that links the deregulation of glutamate, calcium, apoptosis, and the activation of the immune system in schizophrenia patients. All MS data are available in the ProteomeXchange Repository under the identifier PXD015356 and PXD014350.
Collapse
Affiliation(s)
- Erika Velásquez
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-909 , Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry, Institute of Biology , University of Campinas (UNICAMP) , Campinas 13083-970 , Brazil.,Experimental Medicine Research Cluster (EMRC) University of Campinas , Campinas 13083-887 , SP , Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION) , Conselho Nacional de Desenvolvimento Cientı́fico e Tecnológico (CNPq) , São Paulo , Brazil
| | | | - Gabriel Reis Alves Carneiro
- Laboratory of Proteomics, LADETEC, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-598 , Brazil
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy , Ludwig Maximilian University of Munich (LMU) , 80539 Munich , Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy , Ludwig Maximilian University of Munich (LMU) , 80539 Munich , Germany
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-909 , Brazil
| | - Fabio C S Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-909 , Brazil.,Laboratory of Proteomics, LADETEC, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-598 , Brazil
| |
Collapse
|
16
|
López E, Lee-Rivera I, Alvarez-Arce A, López-Colomé AM. Thrombin induces Ca 2+-dependent glutamate release from RPE cells mediated by PLC/PKC and reverse Na +/Ca 2+ exchange. Mol Vis 2019; 25:546-558. [PMID: 31673221 PMCID: PMC6798708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 10/03/2019] [Indexed: 11/01/2022] Open
Abstract
Purpose We analyzed the molecular mechanisms leading to glutamate release from rat primary cultures of RPE cells, under isosmotic conditions. Thrombin has been shown to stimulate glutamate release from astrocytes and retinal glia; however, the effect of thrombin on glutamate release from RPE cells has not been examined. Our previous work showed that upon the alteration of the blood-retina barrier, the serine protease thrombin could contribute to the transformation, proliferation, and migration of RPE cells. In this condition, elevated extracellular glutamate causes neuronal loss in many retinal disorders, including glaucoma, ischemia, diabetic retinopathy, and inherited photoreceptor degeneration. Methods Primary cultures of rat RPE cells were preloaded with 1 µCi/ml 3H-glutamate in Krebs Ringer Bicarbonate (KRB) buffer for 30 min at 37 °C. Cells were rinsed and super-perfused with 1 ml/min KRB for 15 min. Stable release was reached at the 7th minute, and on the 8th minute, fresh KRB containing stimuli was added. Results This study showed for the first time that thrombin promotes specific, dose-dependent glutamate release from RPE cells, induced by the activation of protease-activated receptor 1 (PAR-1). This effect was found to depend on the Ca2+ increase mediated by the phospholipase C-β (PLC-β) and protein kinase C (PKC) pathways, as well as by the reverse activity of the Na+/Ca2+ exchanger. Conclusions Given the intimate contact of the RPE with the photoreceptor outer segments, diffusion of RPE-released glutamate could contribute to the excitotoxic death of retinal neurons, and the development of thrombin-induced eye pathologies.
Collapse
|
17
|
Frinchi M, Nuzzo D, Scaduto P, Di Carlo M, Massenti MF, Belluardo N, Mudò G. Anti-inflammatory and antioxidant effects of muscarinic acetylcholine receptor (mAChR) activation in the rat hippocampus. Sci Rep 2019; 9:14233. [PMID: 31578381 PMCID: PMC6775129 DOI: 10.1038/s41598-019-50708-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/19/2019] [Indexed: 12/17/2022] Open
Abstract
Recently we found that acute treatment with Oxotremorine (Oxo), a non-selective mAChRs agonist, up-regulates heat shock proteins and activates their transcription factor heat shock factor 1 in the rat hippocampus. Here we aimed to investigate: a) if acute treatment with Oxo may regulate pro-inflammatory or anti-inflammatory cytokines and oxidative stress in the rat hippocampus; b) if chronic restraint stress (CRS) induces inflammatory or oxidative alterations in the hippocampus and whether such alterations may be affected by chronic treatment with Oxo. In the acute experiment, rats were injected with single dose of Oxo (0.4 mg/kg) and sacrificed at 24 h, 48 h and 72 h. In the CRS experiment, the rats were exposed for 21 days to the CRS and then were treated with Oxo (0.2 mg/kg) for further 10 days. The acute Oxo treatment showed an ability to significantly reduce reactive oxygen species (ROS), singlet oxygen (1O2), pro-inflammatory cytokines levels (IL-1β and IL-6) and phosphorylated NF-κB-p65. Acute Oxo treatment also increased superoxide dismutase (SOD)-2 protein levels and stimulated SOD activity. No differences were detected in the anti-inflammatory cytokine levels, including IL-10 and TGF-β1. In the group of rats exposed to the CRS were found increased hippocampal IL-1β and IL-6 levels, together with a reduction of SOD activity level. These changes produced by CRS were counteracted by chronic Oxo treatment. In contrast, the upregulation of ROS and 1O2 levels in the CRS group was not counteracted by chronic Oxo treatment. The results revealed a hippocampal anti-inflammatory and antioxidant effect of Oxo treatment in both basal conditions and anti-inflammatory in the CRS rat model.
Collapse
Affiliation(s)
- Monica Frinchi
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Domenico Nuzzo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146, Palermo, Italy
| | - Pietro Scaduto
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Marta Di Carlo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146, Palermo, Italy
| | - Maria F Massenti
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo, 90134, Palermo, Italy
| | - Natale Belluardo
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Giuseppa Mudò
- Department of Biomedicine, Neurosciences and Advanced Diagnostic, div. of Human Physiology, University of Palermo, 90134, Palermo, Italy.
| |
Collapse
|
18
|
Ameliorative effect of imperatorin in chemically induced fibromyalgia: Role of NMDA/NFkB mediated downstream signaling. Biochem Pharmacol 2019; 166:56-69. [DOI: 10.1016/j.bcp.2019.05.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/06/2019] [Indexed: 12/29/2022]
|
19
|
Parra-Damas A, Saura CA. Synapse-to-Nucleus Signaling in Neurodegenerative and Neuropsychiatric Disorders. Biol Psychiatry 2019; 86:87-96. [PMID: 30846302 DOI: 10.1016/j.biopsych.2019.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/18/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023]
Abstract
Synapse-to-nucleus signaling is critical for converting signals received at synapses into transcriptional programs essential for cognition, memory, and emotion. This neuronal mechanism usually involves activity-dependent translocation of synaptonuclear factors from synapses to the nucleus resulting in regulation of transcriptional programs underlying synaptic plasticity. Acting as synapse-to-nucleus messengers, amyloid precursor protein intracellular domain associated-1 protein, cAMP response element binding protein (CREB)-regulated transcription coactivator-1, Jacob, nuclear factor kappa-light-chain-enhancer of activated B cells, RING finger protein 10, and SH3 and multiple ankyrin repeat domains 3 play essential roles in synapse remodeling and plasticity, which are considered the cellular basis of memory. Other synaptic proteins, such as extracellular signal-regulated kinase, calcium/calmodulin-dependent protein kinase II gamma, and CREB2, translocate from dendrites or cytosol to the nucleus upon synaptic activity, suggesting that they could contribute to synapse-to-nucleus signaling. Notably, some synaptonuclear factors converge on the transcription factor CREB, indicating that CREB signaling is a key hub mediating integration of synaptic signals into transcriptional programs required for neuronal function and plasticity. Although major efforts have been focused on identification and regulatory mechanisms of synaptonuclear factors, the relevance of synapse-to-nucleus communication in brain physiology and pathology is still unclear. Recent evidence, however, indicates that synaptonuclear factors are implicated in neuropsychiatric, neurodevelopmental, and neurodegenerative disorders, suggesting that uncoupling synaptic activity from nuclear signaling may prompt synapse pathology, contributing to a broad spectrum of brain disorders. This review summarizes current knowledge of synapse-to-nucleus signaling in neuron survival, synaptic function and plasticity, and memory. Finally, we discuss how altered synapse-to-nucleus signaling may lead to memory and emotional disturbances, which is relevant for clinical and therapeutic strategies in neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Arnaldo Parra-Damas
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
20
|
Dresselhaus EC, Meffert MK. Cellular Specificity of NF-κB Function in the Nervous System. Front Immunol 2019; 10:1043. [PMID: 31143184 PMCID: PMC6520659 DOI: 10.3389/fimmu.2019.01043] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/24/2019] [Indexed: 12/17/2022] Open
Abstract
Nuclear Factor Kappa B (NF-κB) is a ubiquitously expressed transcription factor with key functions in a wide array of biological systems. While the role of NF-κB in processes, such as host immunity and oncogenesis has been more clearly defined, an understanding of the basic functions of NF-κB in the nervous system has lagged behind. The vast cell-type heterogeneity within the central nervous system (CNS) and the interplay between cell-type specific roles of NF-κB contributes to the complexity of understanding NF-κB functions in the brain. In this review, we will focus on the emerging understanding of cell-autonomous regulation of NF-κB signaling as well as the non-cell-autonomous functional impacts of NF-κB activation in the mammalian nervous system. We will focus on recent work which is unlocking the pleiotropic roles of NF-κB in neurons and glial cells (including astrocytes and microglia). Normal physiology as well as disorders of the CNS in which NF-κB signaling has been implicated will be discussed with reference to the lens of cell-type specific responses.
Collapse
Affiliation(s)
- Erica C Dresselhaus
- Department of Biological Chemistry and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mollie K Meffert
- Department of Biological Chemistry and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Cespuglio R, Amrouni D, Raymond EF, Bouteille B, Buguet A. Cerebral inducible nitric oxide synthase protein expression in microglia, astrocytes and neurons in Trypanosoma brucei brucei-infected rats. PLoS One 2019; 14:e0215070. [PMID: 30995270 PMCID: PMC6469759 DOI: 10.1371/journal.pone.0215070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/26/2019] [Indexed: 11/18/2022] Open
Abstract
To study the anatomo-biochemical substrates of brain inflammatory processes, Wistar male rats were infected with Trypanosoma brucei brucei. With this reproducible animal model of human African trypanosomiasis, brain cells (astrocytes, microglial cells, neurons) expressing the inducible nitric oxide synthase (iNOS) enzyme were revealed. Immunohistochemistry was achieved for each control and infected animal through eight coronal brain sections taken along the caudorostral axis of the brain (brainstem, cerebellum, diencephalon and telencephalon). Specific markers of astrocytes (anti-glial fibrillary acidic protein), microglial cells (anti-integrin alpha M) or neurons (anti-Neuronal Nuclei) were employed. The iNOS staining was present in neurons, astrocytes and microglial cells, but not in oligodendrocytes. Stained astrocytes and microglial cells resided mainly near the third cavity in the rostral part of brainstem (periaqueductal gray), diencephalon (thalamus and hypothalamus) and basal telencephalon. Stained neurons were scarce in basal telencephalon, contrasting with numerous iNOS-positive neuroglial cells. Contrarily, in dorsal telencephalon (neocortex and hippocampus), iNOS-positive neurons were plentiful, contrasting with the marked paucity of labelled neuroglial (astrocytes and microglial) cells. The dual distribution between iNOS-labelled neuroglial cells and iNOS-labelled neurons is a feature that has never been described before. Functionalities attached to such a divergent distribution are discussed.
Collapse
Affiliation(s)
- Raymond Cespuglio
- Neuroscience Research Centre of Lyon (CRNL), Neurochem, Faculty of Medicine, Claude-Bernard Lyon-1 University, Lyon, France
- Sechenov 1st Moscow State Medical University, Laboratory of Psychiatric Neurobiology, Moscow, Russia
| | - Donia Amrouni
- Neuroscience Research Centre of Lyon (CRNL), Neurochem, Faculty of Medicine, Claude-Bernard Lyon-1 University, Lyon, France
| | - Elizabeth F. Raymond
- Faculty of Medicine, team EA 4171, Claude-Bernard Lyon-1 University, Lyon, France
| | - Bernard Bouteille
- Department of Parasitology, Dupuytren University Hospital, Limoges, France
| | - Alain Buguet
- Malaria Research Unit, UMR 5246 CNRS, Claude-Bernard Lyon-1 University, Villeurbanne, France
| |
Collapse
|
22
|
Jha NK, Jha SK, Kar R, Nand P, Swati K, Goswami VK. Nuclear factor-kappa β as a therapeutic target for Alzheimer's disease. J Neurochem 2019; 150:113-137. [PMID: 30802950 DOI: 10.1111/jnc.14687] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/16/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a typical progressive, chronic neurodegenerative disorder with worldwide prevalence. Its clinical manifestation involves the presence of extracellular plaques and intracellular neurofibrillary tangles (NFTs). NFTs occur in brain tissues as a result of both Aβ agglomeration and Tau phosphorylation. Although there is no known cure for AD, research into possible cures and treatment options continues using cell-cultures and model animals/organisms. The nuclear factor-kappa β (NF-κβ) plays an active role in the progression of AD. Impairment to this signaling module triggers undesirable phenotypic changes such as neuroinflammation, activation of microglia, oxidative stress related complications, and apoptotic cell death. These imbalances further lead to homeostatic abnormalities in the brain or in initial stages of AD essentially pushing normal neurons toward the degeneration process. Interestingly, the role of NF-κβ signaling associated receptor-interacting protein kinase is currently observed in apoptotic and necrotic cell death, and has been reported in brains. Conversely, the NF-κβ signaling pathway has also been reported to be involved in normal brain functioning. This pathway plays a crucial role in maintaining synaptic plasticity and balancing between learning and memory. Since any impairment in the pathways associated with NF-κβ signaling causes altered neuronal dynamics, neurotherapeutics using compounds including, antioxidants, bioflavonoids, and non-steroidal anti-inflammatory drugs against such abnormalities offer possibilities to rectify aberrant excitatory neuronal activity in AD. In this review, we have provided an extensive overview of the crucial role of NF-κβ signaling in normal brain homeostasis. We have also thoroughly outlined several established pathomechanisms associated with NF-κβ pathways in AD, along with their respective therapeutic approaches.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Rohan Kar
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Parma Nand
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Kumari Swati
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Vineet Kumar Goswami
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| |
Collapse
|
23
|
Association of SHMT1, MAZ, ERG, and L3MBTL3 Gene Polymorphisms with Susceptibility to Multiple Sclerosis. Biochem Genet 2018; 57:355-370. [PMID: 30456721 DOI: 10.1007/s10528-018-9894-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is the most common inflammatory and chronic disease of the central nervous system (CNS). A complex interaction between genetic, environmental, and epigenetic factors is involved in the pathogenesis of MS. With the advancement of GWAS, various variants associated with MS have been identified. This study aimed to evaluate the association of single-nucleotide polymorphisms (SNPs) rs4925166 and rs1979277 in the SHMT1, MAZ rs34286592, ERG rs2836425, and L3MBTL3 rs4364506 with MS. In this case-control study, the association of five SNPs in SHMT1, MAZ, ERG, and L3MBTL3 genes with relapsing-remitting MS (RR-MS) was investigated in 190 patients and 200 healthy individuals. Four SNPs including SHMT1 rs4925166, SHMT1 rs1979277, MAZ rs34286592, and L3MBTL3 rs4364506 were genotyped using PCR-RFLP and genotyping of ERG rs2836425 was performed by tetra-primer ARMS PCR. Our findings showed a significant difference in the allelic frequencies for the four SNPs of SHMT1 rs4925166, SHMT1 rs1979277, MAZ rs34286592, and ERG rs2836425, while there were no differences in the allele and genotype frequencies for L3MBTL3 rs4364506. These significant associations were observed for the following genotypes: TT and GG genotypes of SHMT1 rs4925166 (OR 0.47 and 1.90, respectively) genotype GG of SHMT1 rs1979277 (OR 0.63), genotype GG of MAZ rs34286592 (OR 0.61), TC and CC genotypes of ERG rs2836425 (OR 1.89 and 0.50, respectively). Our study highlighted that people who are carrying genotypes including GG (SHMT1 rs4925166) and TC (ERG rs2836425) have the highest susceptibility chance for MS, respectively. However, genotypes TT (SHMT1 rs4925166), CC (ERG rs2836425), GG (MAZ rs34286592), and GG (SHMT1 rs1979277) had the highest negative association (protective effect) with MS, respectively. L3MBTL3 rs4364506 was found neither as a predisposing nor a protective variant.
Collapse
|
24
|
Ruiz-Perera LM, Schneider L, Windmöller BA, Müller J, Greiner JFW, Kaltschmidt C, Kaltschmidt B. NF-κB p65 directs sex-specific neuroprotection in human neurons. Sci Rep 2018; 8:16012. [PMID: 30375448 PMCID: PMC6207661 DOI: 10.1038/s41598-018-34394-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
Abstract
Protection of neurons against oxidative stress is crucial during neuronal development, maintenance and for treating neurodegenerative diseases. However, little is known about the molecular mechanisms underlying sex-specific maturation and survival of neurons. In the present study, we demonstrate NF-κB-p65 mediated neuroprotection in human glutamatergic neurons differentiated from inferior turbinate stem cells (ITSCs) in a sex-dependent manner. We successfully differentiated ITSCs into MAP-2+/NF200+/Synaptophysin+/vGlut2+-glutamatergic neurons in vitro and ex vivo and validated their functionality. TNF-α-dependent NF-κB-p65 activation was accompanied by significant neuroprotection against oxidative stress-induced neuronal death, which was surprisingly higher in neurons from female donors. Accordingly, sex-specific neuroprotection of female neurons was followed by an increased expression of special NF-κB target genes SOD2 and IGF2. Among these, SOD2 is a well known gene protecting cells against oxidative stress resulting in longevity. In addition, IGF2 is known to promote synapse formation and spine maturation, and it has antioxidant and neuroprotective effects against oxidative damage. In conclusion, we show that NF-κB-p65 is a key player in neuroprotection of human neurons, however the protective gene expression program beneath it differs between sexes. Our findings are in accordance with the increasing evidences pointing towards sex-specific differences in risk and severity of neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Janine Müller
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | | | | | - Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany. .,Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.
| |
Collapse
|
25
|
Macht VA, Reagan LP. Chronic stress from adolescence to aging in the prefrontal cortex: A neuroimmune perspective. Front Neuroendocrinol 2018; 49:31-42. [PMID: 29258741 DOI: 10.1016/j.yfrne.2017.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/22/2017] [Accepted: 12/15/2017] [Indexed: 12/21/2022]
Abstract
The development of the organism is a critical variable which influences the magnitude, duration, and reversibility of the effects of chronic stress. Such factors are relevant to the prefrontal cortex (PFC), as this brain region is the last to mature, the first to decline, and is highly stress-sensitive. Therefore, this review will examine the intersection between the nervous system and immune system at glutamatergic synapses in the PFC across three developmental periods: adolescence, adulthood, and aging. Glutamatergic synapses are tightly juxtaposed with microglia and astrocytes, and each of these cell types exhibits their own developmental trajectory. Not only does chronic stress differentially impact each of these cell types across development, but chronic stress also alters intercellular communication within this quad-partite synapse. These observations suggest that developmental shifts in both neural and immune function across neurons, microglia, and astrocytes mediate shifting effects of chronic stress on glutamatergic transmission.
Collapse
Affiliation(s)
- Victoria A Macht
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, United States; University of South Carolina, Department of Psychology, Columbia, SC, United States.
| | - Lawrence P Reagan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC, United States; Wm. Jennings Bryan Dorn VA Medical Center, Columbia, SC, United States
| |
Collapse
|
26
|
Notch signaling and neuronal death in stroke. Prog Neurobiol 2018; 165-167:103-116. [PMID: 29574014 DOI: 10.1016/j.pneurobio.2018.03.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/08/2018] [Accepted: 03/20/2018] [Indexed: 12/18/2022]
Abstract
Ischemic stroke is a leading cause of morbidity and death, with the outcome largely determined by the amount of hypoxia-related neuronal death in the affected brain regions. Cerebral ischemia and hypoxia activate the Notch1 signaling pathway and four prominent interacting pathways (NF-κB, p53, HIF-1α and Pin1) that converge on a conserved DNA-associated nuclear multi-protein complex, which controls the expression of genes that can determine the fate of neurons. When neurons experience a moderate level of ischemic insult, the nuclear multi-protein complex up-regulates adaptive stress response genes encoding proteins that promote neuronal survival, but when ischemia is more severe the nuclear multi-protein complex induces genes encoding proteins that trigger and execute a neuronal death program. We propose that the nuclear multi-protein transcriptional complex is a molecular mediator of neuronal hormesis and a target for therapeutic intervention in stroke.
Collapse
|
27
|
Savarese AM, Lasek AW. Transcriptional Regulators as Targets for Alcohol Pharmacotherapies. Handb Exp Pharmacol 2018; 248:505-533. [PMID: 29594350 PMCID: PMC6242703 DOI: 10.1007/164_2018_101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing brain disease that currently afflicts over 15 million adults in the United States. Despite its prevalence, there are only three FDA-approved medications for AUD treatment, all of which show limited efficacy. Because of their ability to alter expression of a large number of genes, often with great cell-type and brain-region specificity, transcription factors and epigenetic modifiers serve as promising new targets for the development of AUD treatments aimed at the neural circuitry that underlies chronic alcohol abuse. In this chapter, we will discuss transcriptional regulators that can be targeted pharmacologically and have shown some efficacy in attenuating alcohol consumption when targeted. Specifically, the transcription factors cyclic AMP-responsive element binding protein (CREB), peroxisome proliferator-activated receptors (PPARs), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and glucocorticoid receptor (GR), as well as the epigenetic enzymes, the DNA methyltransferases (DNMTs) and histone deacetylases (HDACs), will be discussed.
Collapse
Affiliation(s)
| | - Amy W. Lasek
- Department of Psychiatry, University of Illinois at Chicago,Corresponding author: 1601 West Taylor Street, MC 912, Chicago, IL 60612, Tel: (312) 355-1593,
| |
Collapse
|
28
|
Abstract
Although autism spectrum disorder (ASD) has a strong genetic basis, its etiology is complex, with several genetic factors likely to be involved as well as environmental factors. Immune dysregulation has gained significant attention as a causal mechanism in ASD pathogenesis. ASD has been associated with immune abnormalities in the brain and periphery, including inflammatory disorders and autoimmunity in not only the affected individuals but also their mothers. Prenatal exposure to maternal immune activation (MIA) has been implicated as an environmental risk factor for ASD. In support of this notion, animal models have shown that MIA results in offspring with behavioral, neurological, and immunological abnormalities similar to those observed in ASD. This raises the question of how MIA exposure can lead to ASD in susceptible individuals. Recent evidence points to a potential inflammation pathway linking MIA-associated ASD with the activity of T helper 17 (Th17) lymphocytes and their effector cytokine interleukin-17A (IL-17A). IL-17A has been implicated from human studies and elevated IL-17A levels in the blood have been found to correlate with phenotypic severity in a subset of ASD individuals. In MIA model mice, elevated IL-17A levels also have been observed. Additionally, antibody blockade to inhibit IL-17A signaling was found to prevent ASD-like behaviors in offspring exposed to MIA. Therefore, IL-17A dysregulation may play a causal role in the development of ASD. The source of increased IL-17A in the MIA mouse model was attributed to maternal Th17 cells because genetic removal of the transcription factor RORγt to selectively inhibit Th17 differentiation in pregnant mice was able to prevent ASD-like behaviors in the offspring. Similar to ASD individuals, the MIA-exposed offspring also displayed cortical dysplasia which could be prevented by inhibition of IL-17A signaling in pregnant mice. This finding reveals one possible cellular mechanism through which ASD-related cognitive and behavioral deficits may emerge following maternal inflammation. IL-17A can exert strong effects on cell survival and differentiation and the activity of signal transduction cascades, which can have important consequences during cortical development on neural function. This review examines IL-17A signaling pathways in the context of both immunity and neural function that may contribute to the development of ASD associated with MIA.
Collapse
Affiliation(s)
- Helen Wong
- Institute for Behavioral Genetics, University of Colorado-Boulder, CO 80303, United States; Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO 80303, United States; Linda Crnic Institute, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Charles Hoeffer
- Institute for Behavioral Genetics, University of Colorado-Boulder, CO 80303, United States; Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO 80303, United States; Linda Crnic Institute, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
29
|
Sakamoto K, Okuwaki T, Ushikubo H, Mori A, Nakahara T, Ishii K. Activation inhibitors of nuclear factor kappa B protect neurons against the NMDA-induced damage in the rat retina. J Pharmacol Sci 2017; 135:S1347-8613(17)30162-7. [PMID: 29110956 DOI: 10.1016/j.jphs.2017.09.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/13/2017] [Indexed: 01/08/2023] Open
Abstract
We reported that high-mobility group Box-1 (HMGB1) was involved in excitoneurotoxicity in the retina. HMGB1 is known to activate nuclear factor kappa B (NF-κB). However, the role of NF-κB in excitotoxicity is still controversial. Here, we demonstrated that NF-κB activation induced by NMDA led to the retinal neurotoxicity. Male Sprague-Dawley rats were used, and NMDA (200 nmol/eye) and bovine HMGB1 (15 μg/eye) were intravitreally injected. Triptolide (500 pmol/eye), BAY 11-7082 (500 pmol/eye), and IMD-0354 (7.5 nmol/eye), NF-κB inhibitors, were co-injected with NMDA or HMGB1. Retinal sections were obtained seven days after intravitreal injection. Cell loss in the ganglion cell layer was observed in the HMGB1- and the NMDA-treated retina. All of the NF-κB inhibitors used in this study reduced the damage. BAY 11-7082 reduced the expression of phosphorylated NF-κB 12 h after NMDA injection, upregulation of GFAP immunoreactivity induced by NMDA 12 and 48 h after NMDA injection, and the number of TUNEL-positive cells 48 h after NMDA injection. The results suggest that NF-κB activation is one of the mechanisms of the retinal neuronal death that occurs 48 h after NMDA injection or later. Prevention of NF-kB activation is a candidate for the treatment of retinal neurodegeneration associated with excitotoxicity.
Collapse
Affiliation(s)
- Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan.
| | - Tatsuya Okuwaki
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Hiroko Ushikubo
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Kunio Ishii
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| |
Collapse
|
30
|
Abstract
Immune control is associated with nigrostriatal neuroprotection for Parkinson's disease (PD); though its direct cause and effect relationships have not yet been realized and modulating the immune system for therapeutic gain has been openly discussed. While the pathobiology of PD remains in study, neuroinflammation is thought to speed nigrostriatal degeneration. The neuroinflammatory cascade associated with PD begins with aggregation of misfolded or post-translationally modified α-synuclein (α-syn). Such aggregation results in neuronal cell death and the presence of chronically activated glia (microglia and astroglia), leading to the production of proinflammatory cytokines like tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), IL-6, and enzymes such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and cyclooxygenase-2 (COX-2). These changes in the glial phenotype can affect the central nervous system (CNS) microenvironment by producing a pro-inflammatory milieu that speeds PD pathogenesis. Mucuna pruriens (Mp) is the most popular drug in Ayurveda, the Indian system of medicine. Several reports have suggested that it possesses analgesic, anti-inflammatory, anti-neoplastic, anti-epileptic and anti-microbial activities. Mp contain L-DOPA and ursolic acid which has an anti-inflammatory property. There are very few literatures which show the immunomodulatory activity of Mp in PD, several researchers have tried to work on the immunomodulatory activity of Mp in some other diseases. The results of several studies show that Mp modulate the immune components like TNF-α, IL-6, IFN-λ, IL-1β, iNOS and IL-2 in the CNS. It also modulates the activity of the transcription factor NF-kB which plays an important role in the progression of the PD. Thus, by altering these cytokines or transcription factors, Mp protects or prevents the progression of PD. Thus in this review we try to explore the immunomodulatory activity of Mp in PD.
Collapse
|
31
|
Zhang J, Zhang Z, Xiang J, Cai M, Yu Z, Li X, Wu T, Cai D. Neuroprotective Effects of Echinacoside on Regulating the Stress-Active p38MAPK and NF-κB p52 Signals in the Mice Model of Parkinson's Disease. Neurochem Res 2016; 42:975-985. [PMID: 27981472 DOI: 10.1007/s11064-016-2130-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/26/2016] [Accepted: 11/29/2016] [Indexed: 12/15/2022]
Abstract
Herbal medicines have long been used to treat Parkinson's disease (PD). To systematically analyze the anti-parkinsonian activity of echinacoside (ECH) in a neurotoxic model of PD and provide a future basis for basic and clinical investigations, male C57BL/6 mice were randomized into blank control, PD model and ECH-administration groups. ECH significantly suppressed the dopaminergic neuron loss (P < 0.01) caused by MPTP and maintained dopamine content (P < 0.01) and dopamine metabolite content (P < 0.05) compared with that measured in mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced damage. Additionally, ECH inhibited the activation of microglia and astrocytes in the substantia nigra, which suggested the involvement of neuroinflammation. The relevant cytokines were detected with a Proteome Profiler Array, which confirmed that ECH participated in the regulation of seven cytokines. Given that p38 mitogen-activated protein kinase (p38MAPK) and NF-kappaB (NF-κB) signals are considered to be closely related to neuroninflammation, the gene expression levels of p38MAPK and six NF-κB DNA-binding subunits were assessed. Western blotting analysis showed that both p38MAPK and the NF-κB p52 subunit were upregulated in the MPTP group and that ECH downregulated their expressions. Minocycline was administered as the positive control to inhibit neuroinflammation, and no differences were detected between the minocycline- and ECH-mediated inhibition of the p38MAPK and NF-κB p52 signals. In conclusion, echinacoside is a potential novel orally active compound for regulating neuroinflammation and related signals in Parkinson's disease and may provide a new prospect for clinical treatment.
Collapse
Affiliation(s)
- Jingsi Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhennian Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhonghai Yu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ting Wu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
32
|
Kaltschmidt B, Kaltschmidt C. NF-KappaB in Long-Term Memory and Structural Plasticity in the Adult Mammalian Brain. Front Mol Neurosci 2015; 8:69. [PMID: 26635522 PMCID: PMC4656838 DOI: 10.3389/fnmol.2015.00069] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/30/2015] [Indexed: 11/13/2022] Open
Abstract
The transcription factor nuclear factor kappaB (NF-κB) is a well-known regulator of inflammation, stress, and immune responses as well as cell survival. In the nervous system, NF-κB is one of the crucial components in the molecular switch that converts short- to long-term memory-a process that requires de novo gene expression. Here, the researches published on NF-κB and downstream target genes in mammals will be reviewed, which are necessary for structural plasticity and long-term memory, both under normal and pathological conditions in the brain. Genetic evidence has revealed that NF-κB regulates neuroprotection, neuronal transmission, and long-term memory. In addition, after genetic ablation of all NF-κB subunits, a severe defect in hippocampal adult neurogenesis was observed during aging. Proliferation of neural precursors is increased; however, axon outgrowth, synaptogenesis, and tissue homeostasis of the dentate gyrus are hampered. In this process, the NF-κB target gene PKAcat and other downstream target genes such as Igf2 are critically involved. Therefore, NF-κB activity seems to be crucial in regulating structural plasticity and replenishment of granule cells within the hippocampus throughout the life. In addition to the function of NF-κB in neurons, we will discuss on a neuroinflammatory role of the transcription factor in glia. Finally, a model for NF-κB homeostasis on the molecular level is presented, in order to explain seemingly the contradictory, the friend or foe, role of NF-κB in the nervous system.
Collapse
|
33
|
de la Fuente V, Federman N, Zalcman G, Salles A, Freudenthal R, Romano A. NF-κB transcription factor role in consolidation and reconsolidation of persistent memories. Front Mol Neurosci 2015; 8:50. [PMID: 26441513 PMCID: PMC4563083 DOI: 10.3389/fnmol.2015.00050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/24/2015] [Indexed: 12/16/2022] Open
Abstract
Transcriptional regulation is an important molecular process required for long-term neural plasticity and long-term memory (LTM) formation. Thus, one main interest in molecular neuroscience in the last decades has been the identification of transcription factors that are involved in memory processes. Among them, the nuclear factor κB (NF-κB) family of transcription factors has gained interest due to a significant body of evidence that supports a key role of these proteins in synaptic plasticity and memory. In recent years, the interest was particularly reinforced because NF-κB was characterized as an important regulator of synaptogenesis. This function may be explained by its participation in synapse to nucleus communication, as well as a possible local role at the synapse. This review provides an overview of experimental work obtained in the last years, showing the essential role of this transcription factor in memory processes in different learning tasks in mammals. We focus the review on the consolidation and reconsolidation memory phases as well as on the regulation of immediate-early and late genes by epigenetic mechanisms that determine enduring forms of memories.
Collapse
Affiliation(s)
- Verónica de la Fuente
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Noel Federman
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Gisela Zalcman
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Angeles Salles
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Ramiro Freudenthal
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| | - Arturo Romano
- Laboratorio de Neurobiología de la Memoria, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria Buenos Aires, Argentina
| |
Collapse
|
34
|
Zhang Z, Sun T, Niu JG, He ZQ, Liu Y, Wang F. Amentoflavone protects hippocampal neurons: anti-inflammatory, antioxidative, and antiapoptotic effects. Neural Regen Res 2015; 10:1125-33. [PMID: 26330838 PMCID: PMC4541246 DOI: 10.4103/1673-5374.160109] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2015] [Indexed: 12/01/2022] Open
Abstract
Amentoflavone is a natural biflavone compound with many biological properties, including anti-inflammatory, antioxidative, and neuroprotective effects. We presumed that amentoflavone exerts a neuroprotective effect in epilepsy models. Prior to model establishment, mice were intragastrically administered 25 mg/kg amentoflavone for 3 consecutive days. Amentoflavone effectively prevented pilocarpine-induced epilepsy in a mouse kindling model, suppressed nuclear factor-κB activation and expression, inhibited excessive discharge of hippocampal neurons resulting in a reduction in epileptic seizures, shortened attack time, and diminished loss and apoptosis of hippocampal neurons. Results suggested that amentoflavone protected hippocampal neurons in epilepsy mice via anti-inflammation, antioxidation, and antiapoptosis, and then effectively prevented the occurrence of seizures.
Collapse
Affiliation(s)
- Zhen Zhang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China ; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Tao Sun
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China ; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhen-Quan He
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yang Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Feng Wang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China ; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
35
|
Abstract
Nuclear factor kappa B (NF-κB) is a potent transcription factor highly expressed in the central nervous system (CNS) where it has been shown to be required for multiple behavioral paradigms of learning and memory in both mammalian and invertebrate systems. NF-κB dimers are found in neuronal cell bodies, are also present at synapses, and can participate in the activity-dependent regulation of gene expression in response to excitatory neurotransmission. Multiple serine-directed phosphorylation events are critical in the canonical NF-κB activation pathway, including activation of the IκB kinase complex (IKK) and phosphorylation and degradation of the inhibitor of NF-κB (IκB). In this chapter, we describe methods for immunoprecipitation (IP) of the IKK complex from dissociated cultured murine hippocampal neurons, followed by in vitro kinase assay to evaluate excitatory neurotransmission-induced IKK activation by monitoring phosphorylation of a GST-IκBα substrate. These methods can also be successfully implemented in subcellular-reduced brain preparations, such as biochemically isolated synapses.
Collapse
|
36
|
Lecointre M, Vézier C, Bénard M, Ramdani Y, Dupré N, Brasse-Lagnel C, Henry VJ, Roy V, Marret S, Gonzalez BJ, Jégou S, Leroux-Nicollet I. Age-dependent alterations of the NMDA receptor developmental profile and adult behavior in postnatally ketamine-treated mice. Dev Neurobiol 2014; 75:315-33. [PMID: 25220981 DOI: 10.1002/dneu.22232] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/22/2014] [Accepted: 09/11/2014] [Indexed: 01/13/2023]
Abstract
Ketamine is a NMDA receptor (NMDAR) antagonist used in pediatric anesthesia. Given the role of glutamatergic signaling during brain maturation, we studied the effects of a single ketamine injection (40 mg/kg s.c) in mouse neonates depending on postnatal age at injection (P2, P5, or P10) on cortical NMDAR subunits expression and association with Membrane-Associated Guanylate Kinases PSD95 and SAP102. The effects of ketamine injection at P2, P5, or P10 on motor activity were compared in adulthood. Ketamine increased GluN2A and GluN2B mRNA levels in P2-treated mice without change in proteins, while it decreased GluN2B protein in P10-treated mice without change in mRNA. Ketamine reduced GluN2A mRNA and protein levels in P5-treated mice without change in GluN2B and GluN1. Ketamine affected the GluN2A/PSD95 association regardless of the age at injection, while GluN2B/PSD95 association was enhanced only in P5-treated mice. Microdissection of ketamine-treated mouse cortex showed a decrease in GluN2A mRNA level in superficial layers (I-IV) and an increase in all subunit expressions in deep layers (V-VI) in P5- and P10-treated mice, respectively. Our data suggest that ketamine impairs cortical NMDAR subunit developmental profile and delays the synaptic targeting of GluN2A-enriched NMDAR. Ketamine injection at P2 or P10 resulted in hyperlocomotion in adult male mice in an open field, without change in females. Voluntary running-wheel exercise showed age- and sex-dependent alterations of the mouse activity, especially during the dark phase. Overall, a single neonatal ketamine exposure led to short-term NMDAR cortical developmental profile impairments and long-term motor activity alterations persisting in adulthood.
Collapse
Affiliation(s)
- Maryline Lecointre
- ERI28 "Neovasc", Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Faculty of Medicine and Pharmacy, Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ye J, Yang Z, Li C, Cai M, Zhou D, Zhang Q, Wei Y, Wang T, Liu Y. NF-κB signaling and vesicle transport are correlated with the reactivation of the memory trace of morphine dependence. Diagn Pathol 2014; 9:142. [PMID: 25012590 PMCID: PMC4227096 DOI: 10.1186/1746-1596-9-142] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/01/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Morphine has been widely used as a clinical anesthetic and analgesic. However, abuse of morphine might result in psychological and physiological dependence. Previous studies have indicated that memory mechanisms play critical roles in morphine dependence. METHODS Morphine dependence was established in mice utilizing place preference conditioning (CPP). We observed changes in the methylome and transcriptome of the nucleus accumbens during the reactivation of the memory trace. We also monitored for changes in the methylome and transcriptome of mice that were acutely exposed to morphine. RESULTS We detected 165 and 18 differentially expressed genes (DEGs) and 6 and 24 significant methyl-sensitive cut counting (MSCC) windows in the acute morphine treatment and the CPP model, respectively. The changes in the methylome and transcriptome during the acute treatment were mainly caused by a response to the morphine stimulus; most of the DEGs were correlated with hormone or transcription factor activity regulation. The expression levels of Lcn2 and Hspb1, which participate in the activation of NF-κB, were significantly decreased in the CPP morphine treatment model. Besides, the alternative splicing of the curtailed isoform of Caps1 was significantly increased in the CPP morphine-treated group, and the methylation levels of Arf4, Vapa, and Gga3 were decreased. These genes play critical roles in the regulation of the Golgi network. CONCLUSIONS The current study indicates that NF-κB signaling and vesicular transport are correlated with the reactivation of the memory trace in morphine-dependent mice. The results obtained in our study agree with previous observations and identify additional candidate genes for further research. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1196707364133126.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yun Liu
- Institute of Biomedical Sciences, Fudan University, Shanghai, PR China.
| |
Collapse
|
38
|
Salles A, Romano A, Freudenthal R. Synaptic NF-kappa B pathway in neuronal plasticity and memory. ACTA ACUST UNITED AC 2014; 108:256-62. [PMID: 24854662 DOI: 10.1016/j.jphysparis.2014.05.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/14/2014] [Accepted: 05/09/2014] [Indexed: 10/25/2022]
Abstract
Several transcription factors are present at the synapse, and among these are the Rel-NF-kappa B pathway components. NF-kappa B is a constitutive transcription factor, with a strong presence in the brain of which a considerable part is located in the neuropiles. This localization of the transcription factor, plus evidence pointing to different functions, is what gave place to two general hypotheses for synaptic NF-kappa B: (a) The transcription factor plays a role in the synapse to nucleus communication, and it is retrogradely transported from polarized localizations to regulate gene expression; (b) The transcription factor modulates the synaptic function locally. Evidence indicates that both mechanisms can operate simultaneously; here we will present different possibilities of these hypotheses that are supported by an increasing amount of data. We pay special attention to the local role of the transcription factor at the synapse, and based in the described evidence from different animal models, we propose several processes in which the transcription factor may change the synaptic strength.
Collapse
Affiliation(s)
- Angeles Salles
- Laboratorio de Neurobiología de la Memoria, FBMC, FCEyN, UBA, IFIBYNE, CONICET, 2°piso, pabellón II, Intendente Güiraldez 2160, Ciudad Universitaria, CP 1428, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Arturo Romano
- Laboratorio de Neurobiología de la Memoria, FBMC, FCEyN, UBA, IFIBYNE, CONICET, 2°piso, pabellón II, Intendente Güiraldez 2160, Ciudad Universitaria, CP 1428, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Ramiro Freudenthal
- Laboratorio de Neurobiología de la Memoria, FBMC, FCEyN, UBA, IFIBYNE, CONICET, 2°piso, pabellón II, Intendente Güiraldez 2160, Ciudad Universitaria, CP 1428, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
39
|
Different susceptibility of prefrontal cortex and hippocampus to oxidative stress following chronic social isolation stress. Mol Cell Biochem 2014; 393:43-57. [DOI: 10.1007/s11010-014-2045-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/14/2014] [Indexed: 12/17/2022]
|
40
|
NF-κB mediated regulation of adult hippocampal neurogenesis: relevance to mood disorders and antidepressant activity. BIOMED RESEARCH INTERNATIONAL 2014; 2014:612798. [PMID: 24678511 PMCID: PMC3942292 DOI: 10.1155/2014/612798] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/28/2013] [Indexed: 12/18/2022]
Abstract
Adult hippocampal neurogenesis is a peculiar form of process of neuroplasticity that in recent years has gained great attention for its potential implication in cognition and in emotional behavior in physiological conditions. Moreover, a vast array of experimental studies suggested that adult hippocampal neurogenesis may be altered in various neuropsychiatric disorders, including major depression, where its disregulation may contribute to cognitive impairment and/or emotional aspects associated with those diseases. An intriguing area of interest is the potential influence of drugs on adult neurogenesis. In particular, several psychoactive drugs, including antidepressants, were shown to positively modulate adult hippocampal neurogenesis. Among molecules which could regulate adult hippocampal neurogenesis the NF-κB family of transcription factors has been receiving particular attention from our and other laboratories. Herein we review recent data supporting the involvement of NF-κB signaling pathways in the regulation of adult neurogenesis and in the effects of drugs that are endowed with proneurogenic and antidepressant activity. The potential implications of these findings on our current understanding of the process of adult neurogenesis in physiological and pathological conditions and on the search for novel antidepressants are also discussed.
Collapse
|
41
|
Ouabain activates NFκB through an NMDA signaling pathway in cultured cerebellar cells. Neuropharmacology 2013; 73:327-36. [DOI: 10.1016/j.neuropharm.2013.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 06/01/2013] [Accepted: 06/03/2013] [Indexed: 11/21/2022]
|
42
|
Abstract
Nuclear factor-kappa B (NF-κB) is a ubiquitous transcription factor that regulates immune and cell-survival signaling pathways. NF-κB has been reported to be present in neurons wherein it reportedly responds to immune and toxic stimuli, glutamate, and synaptic activity. However, because the brain contains many cell types, assays specifically measuring neuronal NF-κB activity are difficult to perform and interpret. To address this, we compared NF-κB activity in cultures of primary neocortical neurons, mixed brain cells, and liver cells, employing Western blot of NF-κB subunits, electrophoretic mobility shift assay (EMSA) of nuclear κB DNA binding, reporter assay of κB DNA binding, immunofluorescence of the NF-κB subunit protein p65, quantitative real-time polymerase chain reaction (PCR) of NF-κB-regulated gene expression, and enzyme-linked immunosorbent assay (ELISA) of produced proteins. Assay of p65 showed its constitutive presence in cytoplasm and nucleus of neurons at levels significantly lower than in mixed brain or liver cells. EMSA and reporter assays showed that constitutive NF-κB activity was nearly absent in neurons. Induced activity was minimal--many fold lower than in other cell types, as measured by phosphorylation and degradation of the inhibitor IκBα, nuclear accumulation of p65, binding to κB DNA consensus sites, NF-κB reporting, or induction of NF-κB-responsive genes. The most efficacious activating stimuli for neurons were the pro-inflammatory cytokines tumor necrosis factor α (TNFα) and interleukin-beta (IL-β). Neuronal NF-κB was not responsive to glutamate in most assays, and it was also unresponsive to hydrogen peroxide, lipopolysaccharide, norepinephrine, ATP, phorbol ester, and nerve growth factor. The chemokine gene transcripts CCL2, CXCL1, and CXCL10 were strongly induced via NF-κB activation by TNFα in neurons, but many candidate responsive genes were not, including the neuroprotective genes SOD2 and Bcl-xL. Importantly, the level of induced neuronal NF-κB activity in response to TNFα or any other stimulus was lower than the level of constitutive activity in non-neuronal cells, calling into question the functional significance of neuronal NF-κB activity.
Collapse
|
43
|
Nitric oxide donors as neuroprotective agents after an ischemic stroke-related inflammatory reaction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:297357. [PMID: 23691263 PMCID: PMC3649699 DOI: 10.1155/2013/297357] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/17/2022]
Abstract
Cerebral ischemia initiates a cascade of detrimental events including glutamate-associated excitotoxicity, intracellular calcium accumulation, formation of Reactive oxygen species (ROS), membrane lipid degradation, and DNA damage, which lead to the disruption of cellular homeostasis and structural damage of ischemic brain tissue. Cerebral ischemia also triggers acute inflammation, which exacerbates primary brain damage. Therefore, reducing oxidative stress (OS) and downregulating the inflammatory response are options that merit consideration as potential therapeutic targets for ischemic stroke. Consequently, agents capable of modulating both elements will constitute promising therapeutic solutions because clinically effective neuroprotectants have not yet been discovered and no specific therapy for stroke is available to date. Because of their ability to modulate both oxidative stress and the inflammatory response, much attention has been focused on the role of nitric oxide donors (NOD) as neuroprotective agents in the pathophysiology of cerebral ischemia-reperfusion injury. Given their short therapeutic window, NOD appears to be appropriate for use during neurosurgical procedures involving transient arterial occlusions, or in very early treatment of acute ischemic stroke, and also possibly as complementary treatment for neurodegenerative diseases such as Parkinson or Alzheimer, where oxidative stress is an important promoter of damage. In the present paper, we focus on the role of NOD as possible neuroprotective therapeutic agents for ischemia/reperfusion treatment.
Collapse
|
44
|
You WC, Wang CX, Pan YX, Zhang X, Zhou XM, Zhang XS, Shi JX, Zhou ML. Activation of nuclear factor-κB in the brain after experimental subarachnoid hemorrhage and its potential role in delayed brain injury. PLoS One 2013; 8:e60290. [PMID: 23536907 PMCID: PMC3607578 DOI: 10.1371/journal.pone.0060290] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 02/25/2013] [Indexed: 01/20/2023] Open
Abstract
It has been reported that inflammation is involved in brain injury after subarachnoid hemorrhage (SAH). Nuclear factor-κB (NF-κB) is a key transcriptional regulator of inflammatory genes. Here, we used pyrrolidine dithiocarbamate(PDTC), an inhibitor of NF-κB, through intracisternal injection to study the role of NF-κB in delayed brain injury after SAH. A total of 55 rabbits were randomly divided into five groups: the control group; the SAH groups including Day-3, 5, and 7 SAH groups (the rabbits in these groups were sacrificed at 3, 5, 7 days after SAH, respectively); and the PDTC group (n = 11 for each group). Electrophoretic mobility shift assay (EMSA) was performed to detect NF-κB DNA-binding activity. The mRNA levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and intercellular adhesion molecule (ICAM)-1 were evaluated by RT-PCR analysis. Deoxyribonucleic acid fragmentation was detected by TUNEL and p65 immunoactivity was assessed by immunohistochemistry. Our results showed the activation of NF-κB after SAH, especially at day 3 and 5. The activated p65 was detected in neurons. NF-κB DNA-binding activity was suppressed by intracisternal administration of PDTC. Increased levels of the TNF-α, IL-1β, and ICAM-1 mRNA were found in the brain at day 5 after SAH, and which were suppressed in the PDTC group. The number of TUNEL-positive cells also decreased significantly in the PDTC group compared with that in the Day-5 SAH group. These results demonstrated that the activated NF-κB in neurons after SAH plays an important role in regulating the expressions of inflammatory genes in the brain, and ultimately contributes to delayed brain injury.
Collapse
Affiliation(s)
- Wan-Chun You
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chun-xi Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yun-xi Pan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiao-ming Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiang-sheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ji-xin Shi
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Meng-liang Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
45
|
Abstract
Glu (glutamate), the excitatory transmitter at the main signalling pathway in the retina, is critically involved in changes in the protein repertoire through the activation of signalling cascades, which regulate protein synthesis at transcriptional and translational levels. Activity-dependent differential gene expression by Glu is related to the activation of ionotropic and metabotropic Glu receptors; however, recent findings suggest the involvement of Na+-dependent Glu transporters in this process. Within the retina, Glu uptake is aimed at the replenishment of the releasable pool, and for the prevention of excitotoxicity and is carried mainly by the GLAST/EAAT-1 (Na+-dependent glutamate/aspartate transporter/excitatory amino acids transporter-1) located in Müller radial glia. Based on the previous work showing the alteration of GLAST expression induced by Glu, the present work investigates the involvement of GLAST signalling in the regulation of protein synthesis in Müller cells. To this end, we explored the effect of D-Asp (D-aspartate) on Ser-2448 mTOR (mammalian target of rapamycin) phosphorylation in primary cultures of chick Müller glia. The results showed that D-Asp transport induces the time- and dose-dependent phosphorylation of mTOR, mimicked by the transportable GLAST inhibitor THA (threo-β-hydroxyaspartate). Signalling leading to mTOR phosphorylation includes Ca2+ influx, the activation of p60src, phosphatidylinositol 3-kinase, protein kinase B, mTOR and p70S6K. Interestingly, GLAST activity promoted AP-1 (activator protein-1) binding to DNA, supporting a function for transporter signalling in retinal long-term responses. These results add a novel receptor-independent pathway for Glu signalling in Müller glia, and further strengthen the critical involvement of these cells in the regulation of glutamatergic transmission in the retina.
Collapse
|
46
|
Yang CH, Liu XM, Si JJ, Shi HS, Xue YX, Liu JF, Luo YX, Chen C, Li P, Yang JL, Wu P, Lu L. Role of IKK/NF-κB signaling in extinction of conditioned place aversion memory in rats. PLoS One 2012; 7:e39696. [PMID: 22761874 PMCID: PMC3383688 DOI: 10.1371/journal.pone.0039696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/29/2012] [Indexed: 11/19/2022] Open
Abstract
The inhibitor κB protein kinase/nuclear factor κB (IKK/NF-κB) signaling pathway is critical for synaptic plasticity. However, the role of IKK/NF-κB in drug withdrawal-associated conditioned place aversion (CPA) memory is unknown. Here, we showed that inhibition of IKK/NF-κB by sulphasalazine (SSZ; 10 mM, i.c.v.) selectively blocked the extinction but not acquisition or expression of morphine-induced CPA in rats. The blockade of CPA extinction induced by SSZ was abolished by sodium butyrate, an inhibitor of histone deacetylase. Thus, the IKK/NF-κB signaling pathway might play a critical role in the extinction of morphine-induced CPA in rats and might be a potential pharmacotherapy target for opiate addiction.
Collapse
Affiliation(s)
- Cheng-Hao Yang
- Tianjin Medical University, Tianjin, China
- Tianjin Institute of Mental Health, Tianjin Mental Health Center, Tianjin, China
| | - Xiang-Ming Liu
- Department of Thoracic Oncology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ji-Jian Si
- Tianjin Medical University, Tianjin, China
- Tianjin Institute of Mental Health, Tianjin Mental Health Center, Tianjin, China
| | - Hai-Shui Shi
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jian-Feng Liu
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yi-Xiao Luo
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Chen Chen
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Peng Li
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jian-Li Yang
- Tianjin Institute of Mental Health, Tianjin Mental Health Center, Tianjin, China
| | - Ping Wu
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China
| |
Collapse
|
47
|
Rehni AK, Singh N. Ammonium pyrrolidine dithiocarbamate and RS 102895 attenuate opioid withdrawal in vivo and in vitro. Psychopharmacology (Berl) 2012; 220:427-38. [PMID: 21931991 DOI: 10.1007/s00213-011-2489-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 09/05/2011] [Indexed: 01/25/2023]
Abstract
RATIONALE Recently, nuclear factor kappa B is indicated in the precipitation of opioid withdrawal syndrome. NF-κB activation is noted to control the transcription and biochemical activation of chemokines. Opioid receptor activation-linked chemokine stimulation is reported to mediate certain effects produced by prolonged opioid treatment. Ammonium pyrrolidine dithiocarbamate (APD) and RS 102895 are relatively selective inhibitors of NF-κB and C-C chemokine receptor 2, respectively. OBJECTIVES The present study investigates the effect of APD and RS 102895 on morphine withdrawal signs in vitro and in vivo. MATERIALS AND METHODS Morphine was administered twice daily for 5 days, following which a single day 6 injection of naloxone (8 mg/kg, i.p.) precipitated opioid withdrawal syndrome in mice. Withdrawal syndrome was quantitatively assessed in terms of withdrawal severity score and the frequency of jumping, rearing, fore paw licking and circling. Naloxone-induced contraction in morphine-withdrawn isolated rat ileum was employed as an in vitro model. An isobolographic study design was employed in the two models to assess potential synergistic activity between APD and RS 102895. RESULTS APD and RS 102895 dose-dependently attenuated naloxone-induced morphine withdrawal syndrome both in vivo and in vitro. APD was also observed to exert a synergistic interaction with RS 102895. CONCLUSIONS It is concluded that APD and RS 102895 attenuate morphine withdrawal signs possibly by a NF-κB and C-C chemokine receptor 2 activation pathway-linked mechanisms potentially in an interdependent manner.
Collapse
Affiliation(s)
- Ashish K Rehni
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, India
| | | |
Collapse
|
48
|
Karpova A, Bär J, Kreutz MR. Long-distance signaling from synapse to nucleus via protein messengers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:355-76. [PMID: 22351064 DOI: 10.1007/978-3-7091-0932-8_16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The communication between synapses and the cell nucleus has attracted considerable interest for many years. This interest is largely fueled by the idea that synapse-to-nucleus signaling might specifically induce the expression of genes that make long-term memory "stick." However, despite many years of research, it is still essentially unclear how synaptic signals are conveyed to the nucleus, and it remains to a large degree enigmatic how activity-induced gene expression feeds back to synaptic function. In this chapter, we will focus on the activity-dependent synapto-nuclear trafficking of protein messengers and discuss the underlying mechanisms of their retrograde transport and their supposed functional role in neuronal plasticity.
Collapse
Affiliation(s)
- Anna Karpova
- PG Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestr.6, 39118 Magdeburg, Germany
| | | | | |
Collapse
|
49
|
Consequences of early life MK-801 administration: long-term behavioural effects and relevance to schizophrenia research. Behav Brain Res 2011; 227:276-86. [PMID: 22085878 DOI: 10.1016/j.bbr.2011.10.052] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 10/31/2011] [Indexed: 12/13/2022]
Abstract
Animal models contribute significantly to advancing the understanding of schizophrenia neurobiology, in addition to being an important tool for the screening of antipsychotic potential of new compounds. However, the entire spectrum or all the symptoms manifested in schizophrenia cannot be straightforwardly reproduced in animals due to the complexity of the disorder, difference in mental capacities and behaviours, and the ability to quantify or measure the changes. Blockade of the NMDA receptor by the use of MK-801, a non-competitive NMDA receptor antagonist, during the early postnatal period has been proposed to be an experimental model which induces behavioural changes that mimic several aspects of the disorder. The long term behavioural profile arising from this early life manipulation is reviewed herein, with a specific focus on behaviours relevant to a schizophrenia-like condition. Some of the reported neurochemical changes are also compiled. Although this method may be suitable to model some aspects of schizophrenia in rodents, there are unmet areas which need to be addressed, notably the characterisation of its predictive value.
Collapse
|
50
|
Kawamoto EM, Lima LS, Munhoz CD, Yshii LM, Kinoshita PF, Amara FG, Pestana RRF, Orellana AMM, Cipolla-Neto J, Britto LRG, Avellar MCW, Rossoni LV, Scavone C. Influence of N-methyl-D-aspartate receptors on ouabain activation of nuclear factor-κB in the rat hippocampus. J Neurosci Res 2011; 90:213-28. [PMID: 22006678 DOI: 10.1002/jnr.22745] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 06/25/2011] [Accepted: 06/27/2011] [Indexed: 02/06/2023]
Abstract
It has been shown that ouabain (OUA) can activate the Na,K-ATPase complex and mediate intracellular signaling in the central nervous system (CNS). Inflammatory stimulus increases glutamatergic transmission, especially at N-methyl-D-aspartate (NMDA) receptors, which are usually coupled to the activation of nitric oxide synthase (NOS). Nuclear factor-κB (NF-κB) activation modulates the expression of genes involved in development, plasticity, and inflammation. The present work investigated the effects of OUA on NF-κB binding activity in rat hippocampus and the influence of this OUA-Na,K-ATPase signaling cascade in NMDA-mediated NF-κB activation. The findings presented here are the first report indicating that intrahippocampal administration of OUA, in a concentration that did not alter Na,K-ATPase or NOS activity, induced an activation of NF-κB, leading to increases in brain-derived neurotrophic factor (Bdnf), inducible NOS (iNos), tumor necrosis factor-α (Tnf-α), and B-cell leukemia/lymphoma 2 (Bcl2) mRNA levels. This response was not linked to any significant signs of neurodegeneration as showed via Fluoro-Jade B and Nissl stain. Intrahippocampal administration of NMDA induced NF-κB activation and increased NOS and α(2/3) -Na,K-ATPase activities. NMDA treatment further increased OUA-induced NF-κB activation, which was partially blocked by MK-801, an antagonist of NMDA receptor. These results suggest that OUA-induced NF-κB activation is at least in part dependent on Na,K-ATPase modulatory action of NMDA receptor in hippocampus. The interaction of these signaling pathways could be associated with biological mechanisms that may underlie the basal homeostatic state linked to the inflammatory signaling cascade in the brain.
Collapse
Affiliation(s)
- E M Kawamoto
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|