1
|
Fan Y, Luan X, Wang X, Li H, Zhao H, Li S, Li X, Qiu Z. Exploring the association between BDNF related signaling pathways and depression: A literature review. Brain Res Bull 2025; 220:111143. [PMID: 39608613 DOI: 10.1016/j.brainresbull.2024.111143] [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] [Received: 09/13/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
Depression is a debilitating mental disease that inflicts significant harm upon individuals and society, yet effective treatment options remain elusive. At present, the pathogenesis of multiple depression is not fully clear, but its occurrence can be related to biological or environmental pathways, among which Brain-derived neurotrophic factor (BDNF) can unequivocally act on two downstream receptors, tyrosine kinase receptor (TrkB) and the p75 neurotrophin receptor (p75NTR), then affect the related signal pathways, affecting the occurrence and development of depression. Accumulating studies have revealed that BDNF-related pathways are critical in the pathophysiology of depression, and their interaction can further influence the efficacy of depression treatment. In this review, we mainly summarized the signaling pathways associated with BDNF and classified them according to different receptors and related molecules, providing promising insights and future directions in the treatment of depression.
Collapse
Affiliation(s)
- Yuchen Fan
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Xinchi Luan
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Xuezhe Wang
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Hongchi Li
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Hongjiao Zhao
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Sheng Li
- Qingdao medical college, Qingdao University, Qingdao, Shandong, China.
| | - Xiaoxuan Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
2
|
Lipsky RH, Witkin JM, Shafique H, Smith JL, Cerne R, Marini AM. Traumatic brain injury: molecular biomarkers, genetics, secondary consequences, and medical management. Front Neurosci 2024; 18:1446076. [PMID: 39450122 PMCID: PMC11500614 DOI: 10.3389/fnins.2024.1446076] [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: 06/08/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Traumatic brain injury (TBI) has reached epidemic proportions worldwide. The consequences of TBI can be severe even with repetitive mild trauma. If death and coma are avoided, the consequences of TBI in the long term typically involve dizziness, sleep disturbances, headache, seizures, cognitive impairment, focal deficits, depression, and anxiety. The severity of brain injury is a significant predictor of outcome. However, the heterogenous nature of the injury makes prognosis difficult. The present review of the literature focuses on the genetics of TBI including genome wide (GWAS) data and candidate gene associations, among them brain-derived neurotrophic factor (BDNF) with TBI and development of post-traumatic epilepsy (PTE). Molecular biomarkers of TBI are also discussed with a focus on proteins and the inflammatory protein IL1-β. The secondary medical sequela to TBI of cognitive impairment, PTE, headache and risk for neurodegenerative disorders is also discussed. This overview of TBI concludes with a review and discussion of the medical management of TBI and the medicines used for and being developed at the preclinical and clinical stages for the treatment of TBI and its host of life-debilitating symptoms.
Collapse
Affiliation(s)
- Robert H. Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jeffrey M. Witkin
- Laboratory of Antiepileptic Drug Discovery Ascension St. Vincent Hospital, Indianapolis, IN, United States
- Departments of Neuroscience and Trauma Research Ascension St. Vincent Hospital, Indianapolis, IN, United States
| | - Hana Shafique
- Duke University School of Medicine, Durham, NC, United States
| | - Jodi L. Smith
- Laboratory of Antiepileptic Drug Discovery Ascension St. Vincent Hospital, Indianapolis, IN, United States
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery Ascension St. Vincent Hospital, Indianapolis, IN, United States
| | - Ann M. Marini
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
3
|
McNearney TA, Westlund KN. Pluripotential GluN1 (NMDA NR1): Functional Significance in Cellular Nuclei in Pain/Nociception. Int J Mol Sci 2023; 24:13196. [PMID: 37686003 PMCID: PMC10488196 DOI: 10.3390/ijms241713196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The N-methyl-D-aspartate (NMDA) glutamate receptors function as plasma membrane ionic channels and take part in very tightly controlled cellular processes activating neurogenic and inflammatory pathways. In particular, the NR1 subunit (new terminology: GluN1) is required for many neuronal and non-neuronal cell functions, including plasticity, survival, and differentiation. Physiologic levels of glutamate agonists and NMDA receptor activation are required for normal neuronal functions such as neuronal development, learning, and memory. When glutamate receptor agonists are present in excess, binding to NMDA receptors produces neuronal/CNS/PNS long-term potentiation, conditions of acute pain, ongoing severe intractable pain, and potential excitotoxicity and pathology. The GluNR1 subunit (116 kD) is necessary as the anchor component directing ion channel heterodimer formation, cellular trafficking, and the nuclear localization that directs functionally specific heterodimer formation, cellular trafficking, and nuclear functions. Emerging studies report the relevance of GluN1 subunit composition and specifically that nuclear GluN1 has major physiologic potential in tissue and/or subnuclear functioning assignments. The shift of the GluN1 subunit from a surface cell membrane to nuclear localization assigns the GluN1 promoter immediate early gene behavior with access to nuclear and potentially nucleolar functions. The present narrative review addresses the nuclear translocation of GluN1, focusing particularly on examples of the role of GluN1 in nociceptive processes.
Collapse
Affiliation(s)
- Terry A. McNearney
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch Galveston, Galveston, TX 77555-1043, USA;
- Department of Internal Medicine, University of Texas Medical Branch Galveston, Galveston, TX 77555-1043, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555-1043, USA
| | - Karin N. Westlund
- Department of Anesthesiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
- Biomedical Laboratory Research & Development (121F), New Mexico VA Health Care System, Albuquerque, NM 87108-5153, USA
| |
Collapse
|
4
|
Pidoplichko VI, Figueiredo TH, Braga MFM, Pan H, Marini AM. Alpha-linolenic acid enhances the facilitation of GABAergic neurotransmission in the BLA and CA1. Exp Biol Med (Maywood) 2023; 248:596-604. [PMID: 37208920 PMCID: PMC10350796 DOI: 10.1177/15353702231165010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 05/21/2023] Open
Abstract
Hyperexcitability is a major mechanism implicated in several neuropsychiatric disorders, such as organophosphate-induced status epilepticus (SE), primary epilepsy, stroke, spinal cord injury, traumatic brain injury, schizophrenia, and autism spectrum disorders. Underlying mechanisms are diverse, but a functional impairment and loss of GABAergic inhibitory neurons are common features in many of these disorders. While novel therapies abound to correct for the loss of GABAergic inhibitory neurons, it has been difficult at best to improve the activities of daily living for the majority of patients. Alpha-linolenic acid (ALA) is an essential omega-3 polyunsaturated fatty acid found in plants. ALA exerts pleiotropic effects in the brain that attenuate injury in chronic and acute brain disease models. However, the effect of ALA on GABAergic neurotransmission in hyperexcitable brain regions involved in neuropsychiatric disorders, such as the basolateral amygdala (BLA) and CA1 subfield of the hippocampus, is unknown. Administration of a single dose of ALA (1500 nmol/kg) subcutaneously increased the charge transfer of inhibitory postsynaptic potential currents mediated by GABAA receptors in pyramidal neurons by 52% in the BLA and by 92% in the CA1 compared to vehicle animals a day later. Similar results were obtained in pyramidal neurons from the BLA and CA1 when ALA was bath-applied in slices from naïve animals. Importantly, pretreatment with the high-affinity, selective TrkB inhibitor, k252, completely abolished the ALA-induced increase in GABAergic neurotransmission in the BLA and CA1, suggesting a brain-derived neurotrophic factor (BDNF)-mediated mechanism. Addition of mature BDNF (20 ng/mL) significantly increased GABAA receptor inhibitory activity in the BLA and CA1 pyramidal neurons similar to the results obtained with ALA. ALA may be an effective treatment for neuropsychiatric disorders where hyperexcitability is a major feature.
Collapse
Affiliation(s)
- Volodymir I Pidoplichko
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Maria FM Braga
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Hongna Pan
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Ann M Marini
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
5
|
Zulkifli NA, Hassan Z, Mustafa MZ, Azman WNW, Hadie SNH, Ghani N, Mat Zin AA. The potential neuroprotective effects of stingless bee honey. Front Aging Neurosci 2023; 14:1048028. [PMID: 36846103 PMCID: PMC9945235 DOI: 10.3389/fnagi.2022.1048028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/29/2022] [Indexed: 02/11/2023] Open
Abstract
Tropical Meliponini bees produce stingless bee honey (SBH). Studies have shown beneficial properties, including antibacterial, bacteriostatic, anti-inflammatory, neurotherapeutic, neuroprotective, wound, and sunburn healing capabilities. High phenolic acid and flavonoid concentrations offer SBH its benefits. SBH can include flavonoids, phenolic acids, ascorbic acid, tocopherol, organic acids, amino acids, and protein, depending on its botanical and geographic origins. Ursolic acid, p-coumaric acid, and gallic acid may diminish apoptotic signals in neuronal cells, such as nuclear morphological alterations and DNA fragmentation. Antioxidant activity minimizes reactive oxygen species (ROS) formation and lowers oxidative stress, inhibiting inflammation by decreasing enzymes generated during inflammation. Flavonoids in honey reduce neuroinflammation by decreasing proinflammatory cytokine and free radical production. Phytochemical components in honey, such as luteolin and phenylalanine, may aid neurological problems. A dietary amino acid, phenylalanine, may improve memory by functioning on brain-derived neurotrophic factor (BDNF) pathways. Neurotrophin BDNF binds to its major receptor, TrkB, and stimulates downstream signaling cascades, which are crucial for neurogenesis and synaptic plasticity. Through BDNF, SBH can stimulate synaptic plasticity and synaptogenesis, promoting learning and memory. Moreover, BDNF contributes to the adult brain's lasting structural and functional changes during limbic epileptogenesis by acting through the cognate receptor tyrosine receptor kinase B (TrkB). Given the higher antioxidants activity of SBH than the Apis sp. honey, it may be more therapeutically helpful. There is minimal research on SBH's neuroprotective effects, and the related pathways contribute to it is unclear. More research is needed to elucidate the underlying molecular process of SBH on BDNF/TrkB pathways in producing neuroprotective effects.
Collapse
Affiliation(s)
- Nurdarina Ausi Zulkifli
- Department of Pathology, School of Medical Sciences Universiti Sains Malaysia and Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
| | - Mohd Zulkifli Mustafa
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Wan Norlina Wan Azman
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia and Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Siti Nurma Hanim Hadie
- Department of Anatomy, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Nurhafizah Ghani
- Basic and Medical Sciences Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Anani Aila Mat Zin
- Department of Pathology, School of Medical Sciences Universiti Sains Malaysia and Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
6
|
Braga MFM, Juranek J, Eiden LE, Li Z, Figueiredo TH, de Araujo Furtado M, Marini AM. GABAergic circuits of the basolateral amygdala and generation of anxiety after traumatic brain injury. Amino Acids 2022; 54:1229-1249. [PMID: 35798984 DOI: 10.1007/s00726-022-03184-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022]
Abstract
Traumatic brain injury (TBI) has reached epidemic proportions around the world and is a major public health concern in the United States. Approximately 2.8 million individuals sustain a traumatic brain injury and are treated in an Emergency Department yearly in the U.S., and about 50,000 of them die. Persistent symptoms develop in 10-15% of the cases including neuropsychiatric disorders. Anxiety is the second most common neuropsychiatric disorder that develops in those with persistent neuropsychiatric symptoms after TBI. Abnormalities or atrophy in the temporal lobe has been shown in the overwhelming number of TBI cases. The basolateral amygdala (BLA), a temporal lobe structure that consolidates, stores and generates fear and anxiety-based behavioral outputs, is a critical brain region in the anxiety circuitry. In this review, we sought to capture studies that characterized the relationship between human post-traumatic anxiety and structural/functional alterations in the amygdala. We compared the human findings with results obtained with a reproducible mild TBI animal model that demonstrated a direct relationship between the alterations in the BLA and an anxiety-like phenotype. From this analysis, both preliminary insights, and gaps in knowledge, have emerged which may open new directions for the development of rational and more efficacious treatments.
Collapse
Affiliation(s)
- Maria F M Braga
- Department of Anatomy, Physiology and Genetics and Program in Neuroscience, Uniformed Services University of the Health Science School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Jenifer Juranek
- Department of Pediatric Surgery, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77030, USA
| | - Lee E Eiden
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, Bethesda, MD, 20814, USA
| | - Zheng Li
- Section On Synapse Development and Plasticity, National Institute of Mental Health, Intramural Research Program, Bethesda, MD, 20814, USA
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology and Genetics and Program in Neuroscience, Uniformed Services University of the Health Science School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Marcio de Araujo Furtado
- Department of Anatomy, Physiology and Genetics and Program in Neuroscience, Uniformed Services University of the Health Science School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Ann M Marini
- Department of Neurology and Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
7
|
Schiel KA. A beneficial role for elevated extracellular glutamate in Amyotrophic Lateral Sclerosis and cerebral ischemia. Bioessays 2021; 43:e2100127. [PMID: 34585427 DOI: 10.1002/bies.202100127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/06/2022]
Abstract
This hypothesis proposes that increased extracellular glutamate in Amyotrophic Lateral Sclerosis (ALS) and cerebral ischemia, currently viewed as a trigger for excitotoxicity, is actually beneficial as it stimulates the utilization of glutamate as metabolic fuel. Renewed appreciation of glutamate oxidation by ischemic neurons has raised questions regarding the role of extracellular glutamate in ischemia. Is it detrimental, as suggested by excitotoxicity in early in vitro studies, or beneficial, as suggested by its oxidation in later in vivo studies? The answer may depend on the activity of N-methyl-D-aspartate (NMDA) glutamate receptors. Early in vitro procedures co-activated NMDA receptors (NMDARs) containing 2A (GluN2A) and 2B (GluN2B) subunits, an event now believed to trigger excitotoxicity; however, during in vivo ischemia D-serine and zinc molecules are released and these ensure only GluN2B receptors are stimulated. This not only prevents excitotoxicity but also initiates signaling cascades that allow ischemic neurons to import and oxidize glutamate.
Collapse
|
8
|
Leschik J, Lutz B, Gentile A. Stress-Related Dysfunction of Adult Hippocampal Neurogenesis-An Attempt for Understanding Resilience? Int J Mol Sci 2021; 22:7339. [PMID: 34298958 PMCID: PMC8305135 DOI: 10.3390/ijms22147339] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Newborn neurons in the adult hippocampus are regulated by many intrinsic and extrinsic cues. It is well accepted that elevated glucocorticoid levels lead to downregulation of adult neurogenesis, which this review discusses as one reason why psychiatric diseases, such as major depression, develop after long-term stress exposure. In reverse, adult neurogenesis has been suggested to protect against stress-induced major depression, and hence, could serve as a resilience mechanism. In this review, we will summarize current knowledge about the functional relation of adult neurogenesis and stress in health and disease. A special focus will lie on the mechanisms underlying the cascades of events from prolonged high glucocorticoid concentrations to reduced numbers of newborn neurons. In addition to neurotransmitter and neurotrophic factor dysregulation, these mechanisms include immunomodulatory pathways, as well as microbiota changes influencing the gut-brain axis. Finally, we discuss recent findings delineating the role of adult neurogenesis in stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| |
Collapse
|
9
|
Back MK, Ruggieri S, Jacobi E, von Engelhardt J. Amyloid Beta-Mediated Changes in Synaptic Function and Spine Number of Neocortical Neurons Depend on NMDA Receptors. Int J Mol Sci 2021; 22:ijms22126298. [PMID: 34208315 PMCID: PMC8231237 DOI: 10.3390/ijms22126298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Onset and progression of Alzheimer's disease (AD) pathophysiology differs between brain regions. The neocortex, for example, is a brain region that is affected very early during AD. NMDA receptors (NMDARs) are involved in mediating amyloid beta (Aβ) toxicity. NMDAR expression, on the other hand, can be affected by Aβ. We tested whether the high vulnerability of neocortical neurons for Aβ-toxicity may result from specific NMDAR expression profiles or from a particular regulation of NMDAR expression by Aβ. Electrophysiological analyses suggested that pyramidal cells of 6-months-old wildtype mice express mostly GluN1/GluN2A NMDARs. While synaptic NMDAR-mediated currents are unaltered in 5xFAD mice, extrasynaptic NMDARs seem to contain GluN1/GluN2A and GluN1/GluN2A/GluN2B. We used conditional GluN1 and GluN2B knockout mice to investigate whether NMDARs contribute to Aβ-toxicity. Spine number was decreased in pyramidal cells of 5xFAD mice and increased in neurons with 3-week virus-mediated Aβ-overexpression. NMDARs were required for both Aβ-mediated changes in spine number and functional synapses. Thus, our study gives novel insights into the Aβ-mediated regulation of NMDAR expression and the role of NMDARs in Aβ pathophysiology in the somatosensory cortex.
Collapse
|
10
|
Dail ME, Brino MLM, Chambers JE. Effects of novel brain-penetrating oxime acetylcholinesterase reactivators on sarin surrogate-induced changes in rat brain gene expression. J Biochem Mol Toxicol 2021; 35:1-10. [PMID: 33682265 DOI: 10.1002/jbt.22755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/28/2021] [Accepted: 02/24/2021] [Indexed: 11/11/2022]
Abstract
Past assassinations and terrorist attacks demonstrate the need for a more effective antidote against nerve agents and other organophosphates (OP) that cause brain damage through inhibition of acetylcholinesterase (AChE). Our lab has invented a platform of phenoxyalkyl pyridinium oximes (US patent 9,277,937) that demonstrate the ability to cross the blood-brain barrier in in vivo rat tests with a sarin surrogate nitrophenyl isopropyl methylphosphonate (NIMP) and provide evidence of brain penetration by reducing cessation time of seizure-like behaviors, accumulation of glial fibrillary acidic protein (GFAP), and hippocampal neuropathology, as opposed to the currently approved oxime, 2-pyridine aldoxime methyl chloride (2-PAM). Using two of the novel oximes (Oximes 1 and 20), this project examined whether gene expression changes might help explain this protection. Expression changes in the piriform cortex were examined using polymerase chain reaction arrays for inflammatory cytokines and receptors. The hippocampus was examined via quantitative polymerase chain reaction for the expression of immediate-early genes involved in brain repair (Bdnf), increasing neurotoxicity (Fos), and apoptosis control (Jdp2, Bcl2l1, Bcl2l11). In the piriform cortex, NIMP significantly stimulated expression for the macrophage inflammatory proteins CCL4, IL-1A, and IL-1B. Oxime 20 by itself elicited the most changes. When it was given therapeutically post-NIMP, the largest change occurred: a 310-fold repression of the inflammatory cytokine, CCL12. In the hippocampus, NIMP increased the expression of the neurotoxicity marker Fos and decreased the expression of neuroprotective Bdnf and antiapoptotic Bcl2l1. Compared with 2-PAM, Oxime 20 stimulated Bcl2l1 expression more and returned expression closer to the vehicle control values.
Collapse
Affiliation(s)
- Mary E Dail
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, Mississippi State University, College of Veterinary Medicine, Mississippi State, United States, USA
| | - Meghan L M Brino
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, Mississippi State University, College of Veterinary Medicine, Mississippi State, United States, USA
| | - Janice E Chambers
- Department of Comparative Biomedical Sciences, Center for Environmental Health Sciences, Mississippi State University, College of Veterinary Medicine, Mississippi State, United States, USA
| |
Collapse
|
11
|
Robinson B, Gu Q, Kanungo J. Antidepressant Actions of Ketamine: Potential Role of L-Type Calcium Channels. Chem Res Toxicol 2021; 34:1198-1207. [PMID: 33566591 DOI: 10.1021/acs.chemrestox.0c00411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, the United States Food and Drug Administration approved esketamine, the S-enantiomer of ketamine, as a fast-acting therapeutic drug for treatment-resistant depression. Although ketamine is known as an N-methyl-d-aspartate (NMDA) receptor antagonist, the underlying mechanisms of how it elicits an antidepressant effect, specifically at subanesthetic doses, are not clear and remain an advancing field of research interest. On the other hand, high-dose (more than the anesthetic dose) ketamine-induced neurotoxicity in animal models has been reported. There has been progress in understanding the potential pathways involved in ketamine-induced antidepressant effects, some of which include NMDA-receptor antagonism, modulation of voltage-gated calcium channels, and brain-derived neurotrophic factor (BDNF) signaling. Often these pathways have been shown to be linked. Voltage-gated L-type calcium channels have been shown to mediate the rapid-acting antidepressant effects of ketamine, especially involving induction of BDNF synthesis downstream, while BDNF deficiency decreases the expression of L-type calcium channels. This review focuses on the reported studies linking ketamine's rapid-acting antidepressant actions to L-type calcium channels with an objective to present a perspective on the importance of the modulation of intracellular calcium in mediating the effects of subanesthetic (antidepressant) versus high-dose ketamine (anesthetic and potential neurotoxicant), the latter having the ability to reduce intracellular calcium by blocking the calcium-permeable NMDA receptors, which is implicated in potential neurotoxicity.
Collapse
Affiliation(s)
- Bonnie Robinson
- Division of Neurotoxicology, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Qiang Gu
- Division of Neurotoxicology, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Jyotshna Kanungo
- Division of Neurotoxicology, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| |
Collapse
|
12
|
Adenosine and NMDA Receptors Modulate Neuroprotection-Induced NMDA Preconditioning in Mice. J Mol Neurosci 2019; 70:590-599. [DOI: 10.1007/s12031-019-01463-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022]
|
13
|
Miranda M, Morici JF, Zanoni MB, Bekinschtein P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front Cell Neurosci 2019; 13:363. [PMID: 31440144 PMCID: PMC6692714 DOI: 10.3389/fncel.2019.00363] [Citation(s) in RCA: 825] [Impact Index Per Article: 137.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
Brain Derived Neurotrophic Factor (BDNF) is a key molecule involved in plastic changes related to learning and memory. The expression of BDNF is highly regulated, and can lead to great variability in BDNF levels in healthy subjects. Changes in BDNF expression are associated with both normal and pathological aging and also psychiatric disease, in particular in structures important for memory processes such as the hippocampus and parahippocampal areas. Some interventions like exercise or antidepressant administration enhance the expression of BDNF in normal and pathological conditions. In this review, we will describe studies from rodents and humans to bring together research on how BDNF expression is regulated, how this expression changes in the pathological brain and also exciting work on how interventions known to enhance this neurotrophin could have clinical relevance. We propose that, although BDNF may not be a valid biomarker for neurodegenerative/neuropsychiatric diseases because of its disregulation common to many pathological conditions, it could be thought of as a marker that specifically relates to the occurrence and/or progression of the mnemonic symptoms that are common to many pathological conditions.
Collapse
Affiliation(s)
- Magdalena Miranda
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| | - Juan Facundo Morici
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| | - María Belén Zanoni
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| | - Pedro Bekinschtein
- Laboratory of Memory Research and Molecular Cognition, Institute for Cognitive and Translational Neuroscience, Instituto de Neurología Cognitiva, CONICET, Universidad Favaloro, Buenos Aires, Argentina
| |
Collapse
|
14
|
Abstract
Mild environmental stress might have beneficial effects in aging by activating maintenance and repair processes in cells and organs. These beneficial stress effects fit to the concept of hormesis. Prominent stressors acting in a hormetic way are physical exercises, fasting, cold and heat. This review will introduce some toxins, which have been found to induce hormetic responses in animal models of aging research. To highlight the molecular signature of these hormetic effects we will depict signaling pathways affected by low doses of toxins on cellular and organismic level. As prominent examples for signaling pathways involved in both aging processes as well as toxin responses, PI3K/Akt/mTOR- and AMPK-signal transduction will be described in more detail. Due to the striking overlap of signaling pathways mediating toxin induced responses and aging processes we propose considering the ability of low doses of toxins to slow down the rate of aging.
Collapse
|
15
|
Sachana M, Rolaki A, Bal-Price A. Development of the Adverse Outcome Pathway (AOP): Chronic binding of antagonist to N-methyl-d-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities of children. Toxicol Appl Pharmacol 2018; 354:153-175. [PMID: 29524501 PMCID: PMC6095943 DOI: 10.1016/j.taap.2018.02.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 01/06/2023]
Abstract
The Adverse Outcome Pathways (AOPs) are designed to provide mechanistic understanding of complex biological systems and pathways of toxicity that result in adverse outcomes (AOs) relevant to regulatory endpoints. AOP concept captures in a structured way the causal relationships resulting from initial chemical interaction with biological target(s) (molecular initiating event) to an AO manifested in individual organisms and/or populations through a sequential series of key events (KEs), which are cellular, anatomical and/or functional changes in biological processes. An AOP provides the mechanistic detail required to support chemical safety assessment, the development of alternative methods and the implementation of an integrated testing strategy. An example of the AOP relevant to developmental neurotoxicity (DNT) is described here following the requirements of information defined by the OECD Users' Handbook Supplement to the Guidance Document for developing and assessing AOPs. In this AOP, the binding of an antagonist to glutamate receptor N-methyl-d-aspartate (NMDAR) receptor is defined as MIE. This MIE triggers a cascade of cellular KEs including reduction of intracellular calcium levels, reduction of brain derived neurotrophic factor release, neuronal cell death, decreased glutamate presynaptic release and aberrant dendritic morphology. At organ level, the above mentioned KEs lead to decreased synaptogenesis and decreased neuronal network formation and function causing learning and memory deficit at organism level, which is defined as the AO. There are in vitro, in vivo and epidemiological data that support the described KEs and their causative relationships rendering this AOP relevant to DNT evaluation in the context of regulatory purposes.
Collapse
Affiliation(s)
| | | | - Anna Bal-Price
- European Commission, Joint Research Centre, Ispra, Italy.
| |
Collapse
|
16
|
Klass A, Sánchez-Porras R, Santos E. Systematic review of the pharmacological agents that have been tested against spreading depolarizations. J Cereb Blood Flow Metab 2018; 38:1149-1179. [PMID: 29673289 PMCID: PMC6434447 DOI: 10.1177/0271678x18771440] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Spreading depolarization (SD) occurs alongside brain injuries and it can lead to neuronal damage. Therefore, pharmacological modulation of SD can constitute a therapeutic approach to reduce its detrimental effects and to improve the clinical outcome of patients. The major objective of this article was to produce a systematic review of all the drugs that have been tested against SD. Of the substances that have been examined, most have been shown to modulate certain SD characteristics. Only a few have succeeded in significantly inhibiting SD. We present a variety of strategies that have been proposed to overcome the notorious harmfulness and pharmacoresistance of SD. Information on clinically used anesthetic, sedative, hypnotic agents, anti-migraine drugs, anticonvulsants and various other substances have been compiled and reviewed with respect to the efficacy against SD, in order to answer the question of whether a drug at safe doses could be of therapeutic use against SD in humans.
Collapse
Affiliation(s)
- Anna Klass
- Neurosurgery Department, University of Heidelberg, Heidelberg, Germany
| | | | - Edgar Santos
- Neurosurgery Department, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Rubio-Casillas A, Fernández-Guasti A. The dose makes the poison: from glutamate-mediated neurogenesis to neuronal atrophy and depression. Rev Neurosci 2018; 27:599-622. [PMID: 27096778 DOI: 10.1515/revneuro-2015-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/04/2016] [Indexed: 12/21/2022]
Abstract
Experimental evidence has demonstrated that glutamate is an essential factor for neurogenesis, whereas another line of research postulates that excessive glutamatergic neurotransmission is associated with the pathogenesis of depression. The present review shows that such paradox can be explained within the framework of hormesis, defined as biphasic dose responses. Low glutamate levels activate adaptive stress responses that include proteins that protect neurons against more severe stress. Conversely, abnormally high levels of glutamate, resulting from increased release and/or decreased removal, cause neuronal atrophy and depression. The dysregulation of the glutamatergic transmission in depression could be underlined by several factors including a decreased inhibition (γ-aminobutyric acid or serotonin) or an increased excitation (primarily within the glutamatergic system). Experimental evidence shows that the activation of N-methyl-D-aspartate receptor (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPAR) can exert two opposite effects on neurogenesis and neuron survival depending on the synaptic or extrasynaptic concentration. Chronic stress, which usually underlies experimental and clinical depression, enhances glutamate release. This overactivates NMDA receptors (NMDAR) and consequently impairs AMPAR activity. Various studies show that treatment with antidepressants decreases plasma glutamate levels in depressed individuals and regulates glutamate receptors by reducing NMDAR function by decreasing the expression of its subunits and by potentiating AMPAR-mediated transmission. Additionally, it has been shown that chronic treatment with antidepressants having divergent mechanisms of action (including tricyclics, selective serotonin reuptake inhibitors, and ketamine) markedly reduced depolarization-evoked glutamate release in the hippocampus. These data, taken together, suggest that the glutamatergic system could be a final common pathway for antidepressant treatments.
Collapse
|
18
|
Gorkhali R, Huang K, Kirberger M, Yang JJ. Defining potential roles of Pb(2+) in neurotoxicity from a calciomics approach. Metallomics 2017; 8:563-78. [PMID: 27108875 DOI: 10.1039/c6mt00038j] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metal ions play crucial roles in numerous biological processes, facilitating biochemical reactions by binding to various proteins. An increasing body of evidence suggests that neurotoxicity associated with exposure to nonessential metals (e.g., Pb(2+)) involves disruption of synaptic activity, and these observed effects are associated with the ability of Pb(2+) to interfere with Zn(2+) and Ca(2+)-dependent functions. However, the molecular mechanism behind Pb(2+) toxicity remains a topic of debate. In this review, we first discuss potential neuronal Ca(2+) binding protein (CaBP) targets for Pb(2+) such as calmodulin (CaM), synaptotagmin, neuronal calcium sensor-1 (NCS-1), N-methyl-d-aspartate receptor (NMDAR) and family C of G-protein coupled receptors (cGPCRs), and their involvement in Ca(2+)-signalling pathways. We then compare metal binding properties between Ca(2+) and Pb(2+) to understand the structural implications of Pb(2+) binding to CaBPs. Statistical and biophysical studies (e.g., NMR and fluorescence spectroscopy) of Pb(2+) binding are discussed to investigate the molecular mechanism behind Pb(2+) toxicity. These studies identify an opportunistic, allosteric binding of Pb(2+) to CaM, which is distinct from ionic displacement. Together, these data suggest three potential modes of Pb(2+) activity related to molecular and/or neural toxicity: (i) Pb(2+) can occupy Ca(2+)-binding sites, inhibiting the activity of the protein by structural modulation, (ii) Pb(2+) can mimic Ca(2+) in the binding sites, falsely activating the protein and perturbing downstream activities, or (iii) Pb(2+) can bind outside of the Ca(2+)-binding sites, resulting in the allosteric modulation of the protein activity. Moreover, the data further suggest that even low concentrations of Pb(2+) can interfere at multiple points within the neuronal Ca(2+) signalling pathways to cause neurotoxicity.
Collapse
Affiliation(s)
- Rakshya Gorkhali
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| | - Kenneth Huang
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| | - Michael Kirberger
- Department of Chemistry and Physics, Clayton State University, Morrow, GA 30260, USA.
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| |
Collapse
|
19
|
Shin HK, Lee SW, Choi BT. Modulation of neurogenesis via neurotrophic factors in acupuncture treatments for neurological diseases. Biochem Pharmacol 2017; 141:132-142. [DOI: 10.1016/j.bcp.2017.04.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/26/2017] [Indexed: 12/27/2022]
|
20
|
Ghasemi M, Phillips C, Fahimi A, McNerney MW, Salehi A. Mechanisms of action and clinical efficacy of NMDA receptor modulators in mood disorders. Neurosci Biobehav Rev 2017; 80:555-572. [DOI: 10.1016/j.neubiorev.2017.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/23/2017] [Accepted: 07/08/2017] [Indexed: 12/22/2022]
|
21
|
Morris G, Walder K, Carvalho AF, Tye SJ, Lucas K, Berk M, Maes M. The role of hypernitrosylation in the pathogenesis and pathophysiology of neuroprogressive diseases. Neurosci Biobehav Rev 2017; 84:453-469. [PMID: 28789902 DOI: 10.1016/j.neubiorev.2017.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/02/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022]
Abstract
There is a wealth of data indicating that de novo protein S-nitrosylation in general and protein transnitrosylation in particular mediates the bulk of nitric oxide signalling. These processes enable redox sensing and facilitate homeostatic regulation of redox dependent protein signalling, function, stability and trafficking. Increased S-nitrosylation in an environment of increasing oxidative and nitrosative stress (O&NS) is initially a protective mechanism aimed at maintaining protein structure and function. When O&NS becomes severe, mechanisms governing denitrosylation and transnitrosylation break down leading to the pathological state referred to as hypernitrosylation (HN). Such a state has been implicated in the pathogenesis and pathophysiology of several neuropsychiatric and neurodegenerative diseases and we investigate its potential role in the development and maintenance of neuroprogressive disorders. In this paper, we propose a model whereby the hypernitrosylation of a range of functional proteins and enzymes lead to changes in activity which conspire to produce at least some of the core abnormalities contributing to the development and maintenance of pathology in these illnesses.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, SA152LW, Wales, United Kingdom
| | - Ken Walder
- Deakin University, The Centre for Molecular and Medical Research, School of Medicine, P.O. Box 291, Geelong, 3220, Australia
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, 60430-040, Fortaleza, CE, Brazil
| | - Susannah J Tye
- Deakin University, The Centre for Molecular and Medical Research, School of Medicine, P.O. Box 291, Geelong, 3220, Australia; Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, 60430-040, Fortaleza, CE, Brazil; Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia
| | - Kurt Lucas
- Multiphase Chemistry Department, Max Planck Institute for Chemistry, 55128 Mainz, Germany
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia.
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, P.O. Box 281, Geelong, 3220, Australia; Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
22
|
Martucci M, Ostan R, Biondi F, Bellavista E, Fabbri C, Bertarelli C, Salvioli S, Capri M, Franceschi C, Santoro A. Mediterranean diet and inflammaging within the hormesis paradigm. Nutr Rev 2017; 75:442-455. [PMID: 28595318 PMCID: PMC5914347 DOI: 10.1093/nutrit/nux013] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A coherent set of epidemiological data shows that the Mediterranean diet has beneficial effects capable of preventing a variety of age-related diseases in which low-grade, chronic inflammation/inflammaging plays a major role, but the underpinning mechanism(s) is/are still unclear. It is suggested here that the Mediterranean diet can be conceptualized as a form of chronic hormetic stress, similar to what has been proposed regarding calorie restriction, the most thoroughly studied nutritional intervention. Data on the presence in key Mediterranean foods of a variety of compounds capable of exerting hormetic effects are summarized, and the mechanistic role of the nuclear factor erythroid 2 pathway is highlighted. Within this conceptual framework, particular attention has been devoted to the neurohormetic and neuroprotective properties of the Mediterranean diet, as well as to its ability to maintain an optimal balance between pro- and anti-inflammaging. Finally, the European Commission-funded project NU-AGE is discussed because it addresses a number of variables not commonly taken into consideration, such as age, sex, and ethnicity/genetics, that can modulate the hormetic effect of the Mediterranean diet.
Collapse
Affiliation(s)
- Morena Martucci
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Rita Ostan
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Fiammetta Biondi
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Elena Bellavista
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Cristina Fabbri
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Claudia Bertarelli
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Stefano Salvioli
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Miriam Capri
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Claudio Franceschi
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| | - Aurelia Santoro
- M. Martucci, F. Biondi, E. Bellavista, C. Fabbri, C. Bertarelli, S. Salvioli, M. Capri, and A. Santoro are with the Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy. R. Ostan, S. Salvioli, M. Capri, and A. Santoro are with the Interdepartmental Center “L. Galvani” (CIG), University of Bologna, Bologna, Italy. C. Franceschi is with the Institute of Neurological Sciences (IRCCS), Bologna, Italy
| |
Collapse
|
23
|
Leviton A, Allred EN, Yamamoto H, Fichorova RN, Kuban K, O'Shea TM, Dammann O. Antecedents and correlates of blood concentrations of neurotrophic growth factors in very preterm newborns. Cytokine 2017; 94:21-28. [PMID: 28396037 PMCID: PMC5464409 DOI: 10.1016/j.cyto.2017.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/18/2017] [Accepted: 03/31/2017] [Indexed: 11/16/2022]
Abstract
AIM To identify the antecedents and very early correlates of low concentrations of neurotrophic growth factors in the blood of extremely preterm newborns during the first postnatal month. METHODS Using an immunobead assay, we measured the concentrations of neurotrophin 4 (NT4), brain-derived neurotrophic factor (BDNF), and basic fibroblast growth factor (bFGF) in blood spots collected on postnatal days 1 (N=1062), 7 (N=1087), 14 (N=989), 21 (N=940) and 28 (N=880) from infants born before the 28th week of gestation. We then sought the correlates of measurements in the top and bottom quartiles for gestational age and day the specimen was collected. RESULTS The concentrations of 2 neurotrophic proteins, NT4 and BDNF, were low among children delivered for medical (maternal or fetal) indications, and among those who were growth restricted. Children who had top quartile concentrations of NT4, BDNF, and bFGF tended to have elevated concentrations of inflammation-related proteins that day. This pattern persisted for much of the first postnatal month. CONCLUSIONS Delivery for medical indications and fetal growth restriction are associated with a relative paucity of NT4 and BDNF concentrations during the first 24 h after very preterm birth. Elevated blood concentrations of NT4, BDNF, and bFGF tended to co-occur with indicators of systemic inflammation on the same day.
Collapse
Affiliation(s)
- Alan Leviton
- Boston Children's Hospital, and Harvard Medical School, Boston, MA, United States.
| | - Elizabeth N Allred
- Boston Children's Hospital, and Harvard Medical School, Boston, MA, United States
| | | | - Raina N Fichorova
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Karl Kuban
- Boston Medical Center and Boston University, Boston, MA, United States
| | | | - Olaf Dammann
- Tufts University School of Medicine, Boston, MA, United States; Hannover Medical School, Hannover, Germany
| |
Collapse
|
24
|
Can Co-Activation of Nrf2 and Neurotrophic Signaling Pathway Slow Alzheimer's Disease? Int J Mol Sci 2017; 18:ijms18061168. [PMID: 28561773 PMCID: PMC5485992 DOI: 10.3390/ijms18061168] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/22/2017] [Accepted: 05/27/2017] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifaceted disease that is hard to treat by single-modal treatment. AD starts with amyloid peptides, mitochondrial dysfunction, and oxidative stress and later is accompanied with chronic endoplasmic reticulum (ER) stress and autophagy dysfunction, resulting in more complicated pathogenesis. Currently, few treatments can modify the complicated pathogenic progress of AD. Compared to the treatment with exogenous antioxidants, the activation of global antioxidant defense system via Nrf2 looks more promising in attenuating oxidative stress in AD brains. Accompanying the activation of the Nrf2-mediated antioxidant defense system that reduce the AD-causative factor, oxidative stress, it is also necessary to activate the neurotrophic signaling pathway that replaces damaged organelles and molecules with new ones. Thus, the dual actions to activate both the Nrf2 antioxidant system and neurotrophic signaling pathway are expected to provide a better strategy to modify AD pathogenesis. Here, we review the current understanding of AD pathogenesis and neuronal defense systems and discuss a possible way to co-activate the Nrf2 antioxidant system and neurotrophic signaling pathway with the hope of helping to find a better strategy to slow AD.
Collapse
|
25
|
Azogu I, Plamondon H. Blockade of TrkB receptors in the nucleus accumbens prior to heterotypic stress alters corticotropin-releasing hormone (CRH), vesicular glutamate transporter 2 (vGluT2) and glucocorticoid receptor (GR) within the mesolimbic pathway. Horm Behav 2017; 90:98-112. [PMID: 28257759 DOI: 10.1016/j.yhbeh.2017.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/22/2016] [Accepted: 02/22/2017] [Indexed: 12/27/2022]
Abstract
Inhibition of stress-induced elevations in brain-derived neurotrophic factor (BDNF) or its primary receptor tyrosine-related kinase B (TrkB) within the reward pathway may modulate vulnerability to anxiety and mood disorders. The current study examined the role of BDNF/TrkB signaling on biochemistry and behavior under basal conditions and following exposure to a 10-day heterotypic stress paradigm in male rats. Effects of intra-accumbal administration of TrkB antagonist ANA-12 (0.25μg/0.5μl/min) on anxiety, and expression of Trk-B, corticotropin-releasing hormone (CRH), vesicular glutamate transporter 2 (vGluT2) and glucocorticoid receptor (GR) within the mesolimbic pathway were determined. Notably, ANA-12 attenuated anxiety-like behavior in stress rats while increasing anxiety in the non-stress group in the elevated plus maze (EPM). At the neurochemical level, ANA-12 blocked the increased vGluT2 and CRH expressions in the hypothalamic PVN and basolateral amygdala in stress rats, while it enhanced vGluT2 and CRH expressions in non-stress rats. ANA-12 also showed state-dependent effects at the NAc core, attenuating TrkB-ir in non-stress rats while reversing reduced expression in stressed rats. At the cingulate cortex, ANA-12 normalized stress-induced increase in TrkB expression. Notably, ANA-12 showed region-specific effects on GR-ir at the NAc core and shell, with increased GR-ir in non-stress rats, although the drug attenuated stress-induced GR-ir expression only in the core portion of the NAc, while having no impact at the cingulate cortex. Elevated blood CORT levels post-stress was not influenced by ANA-12 treatment. Together, these findings suggest that BDNF-mediated TrkB activation exerts differential impact in regulating emotional response under basal and stress conditions.
Collapse
Affiliation(s)
- Idu Azogu
- Behavioural Neuroscience Group, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, ON, Canada
| | - Helene Plamondon
- Behavioural Neuroscience Group, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, ON, Canada.
| |
Collapse
|
26
|
Barnes AK, Koul-Tiwari R, Garner JM, Geist PA, Datta S. Activation of brain-derived neurotrophic factor-tropomyosin receptor kinase B signaling in the pedunculopontine tegmental nucleus: a novel mechanism for the homeostatic regulation of rapid eye movement sleep. J Neurochem 2017; 141:111-123. [PMID: 28027399 PMCID: PMC5364057 DOI: 10.1111/jnc.13938] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/09/2016] [Accepted: 12/09/2016] [Indexed: 02/04/2023]
Abstract
Rapid eye movement (REM) sleep dysregulation is a symptom of many neuropsychiatric disorders, yet the mechanisms of REM sleep homeostatic regulation are not fully understood. We have shown that, after REM sleep deprivation, the pedunculopontine tegmental nucleus (PPT) plays a critical role in the generation of recovery REM sleep. In this study, we used multidisciplinary techniques to show a causal relationship between brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the PPT and the development of REM sleep homeostatic drive. Rats were randomly assigned to conditions of unrestricted sleep or selective REM sleep deprivation (RSD) with PPT microinjections of vehicle control or a dose of a TrkB receptor inhibitor (2, 3, or 4 nmol K252a or 4 nmol ANA-12). On experimental days, rats received PPT microinjections and their sleep-wake physiological signals were recorded for 3 or 6 h, during which selective RSD was performed in the first 3 h. At the end of all 3 h recordings, rats were killed and the PPT was dissected out for BDNF quantification. Our results show that K252a and ANA-12 dose-dependently reduced the homeostatic responses to selective RSD. Specifically, TrkB receptor inhibition reduced REM sleep homeostatic drive and limited REM sleep rebound. There was also a dose-dependent suppression of PPT BDNF up-regulation, and regression analysis revealed a significant positive relationship between REM sleep homeostatic drive and the level of PPT BDNF expression. These data provide the first direct evidence that activation of BDNF-TrkB signaling in the PPT is a critical step for the development of REM sleep homeostatic drive.
Collapse
Affiliation(s)
- Abigail K Barnes
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Richa Koul-Tiwari
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Jennifer M Garner
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Phillip A Geist
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Subimal Datta
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA.,Program in Comparative and Experimental Medicine, The University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
27
|
Liu L, Huang W, Wang J, Song H, Cen J, Ji B. Anthraquinone derivative exerted hormetic effect on the apoptosis in oxygen-glucose deprivation-induced PC12 cells via ERK and Akt activated Nrf2/HO-1 signaling pathway. Chem Biol Interact 2016; 262:1-11. [PMID: 27923643 DOI: 10.1016/j.cbi.2016.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/24/2016] [Accepted: 12/02/2016] [Indexed: 12/13/2022]
Abstract
There were accumulated evidences that agents may attenuate neurological disorders through a hormetic effect. This study was designed to investigate hormetic effect of BME on the oxygen-glucose deprivation (OGD)-induced mitochondrial apoptosis in NGF-differentiated PC12 cells. The effect of BME on the intracellular reactive oxygen species (iROS) formation and pro-survival signals mediated by ERK and Akt as well as transcription factor nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) pathways was also determined. The present results showed that, at low concentrations, pretreatment with BME triggered stress response by causing ROS production, then, activated survival-promoting signals via ERK and Akt activated Nrf2/HO-1 signaling pathway, resulting in decrease in cytotoxicity induced by the OGD. It may be accepted that mild pretreatment with BME stimulated transient and moderate ROS production, but activated hormetic signals and induced stress responsive genes. In contrast, high concentrations of BME displayed toxic action due to massive ROS production. These results suggested that the effect of BME on the OGD-induced PC12 cells may be hormetic mechanism including induction of oxidative stress and subsequent activation of stress response gene expression.
Collapse
Affiliation(s)
- Lu Liu
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Weiwei Huang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Huiling Song
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China.
| | - Biansheng Ji
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China.
| |
Collapse
|
28
|
Chlorella sorokiniana Extract Improves Short-Term Memory in Rats. Molecules 2016; 21:molecules21101311. [PMID: 27689989 PMCID: PMC6274193 DOI: 10.3390/molecules21101311] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 01/28/2023] Open
Abstract
Increasing evidence shows that eukaryotic microalgae and, in particular, the green microalga Chlorella, can be used as natural sources to obtain a whole variety of compounds, such as omega (ω)-3 and ω-6 polyunsatured fatty acids (PUFAs). Although either beneficial or toxic effects of Chlorella sorokiniana have been mainly attributed to its specific ω-3 and ω-6 PUFAs content, the underlying molecular pathways remain to be elucidated yet. Here, we investigate the effects of an acute oral administration of a lipid extract of Chlorella sorokiniana, containing mainly ω-3 and ω-6 PUFAs, on cognitive, emotional and social behaviour in rats, analysing possible underlying neurochemical alterations. Our results showed improved short-term memory in Chlorella sorokiniana-treated rats compared to controls, without any differences in exploratory performance, locomotor activity, anxiety profile and depressive-like behaviour. On the other hand, while the social behaviour of Chlorella sorokiniana-treated animals was significantly decreased, no effects on aggressivity were observed. Neurochemical investigations showed region-specific effects, consisting in an elevation of noradrenaline (NA) and serotonin (5-HT) content in hippocampus, but not in the prefrontal cortex and striatum. In conclusion, our results point towards a beneficial effect of Chlorella sorokiniana extract on short-term memory, but also highlight the need of caution in the use of this natural supplement due to its possible masked toxic effects.
Collapse
|
29
|
Preconditioning is hormesis part I: Documentation, dose-response features and mechanistic foundations. Pharmacol Res 2016; 110:242-264. [DOI: 10.1016/j.phrs.2015.12.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
|
30
|
Preconditioning is hormesis part II: How the conditioning dose mediates protection: Dose optimization within temporal and mechanistic frameworks. Pharmacol Res 2016; 110:265-275. [DOI: 10.1016/j.phrs.2015.12.020] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 01/02/2023]
|
31
|
Protective effect of liquiritigenin on depressive-like behavior in mice after lipopolysaccharide administration. Psychiatry Res 2016; 240:131-136. [PMID: 27107388 DOI: 10.1016/j.psychres.2016.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/27/2016] [Accepted: 04/01/2016] [Indexed: 12/16/2022]
Abstract
Liquiritigenin (Liq), the main active ingredient of traditional Chinese medicine licorice, possesses anti-inflammatory and neuroprotective properties. The current investigation was designed to explore whether liquiritigenin could relieve lipopolysaccharide (LPS)-induced depression-like behavior in mice and the underlying mechanism. Liquiritigenin (7.5mg/kg, 15mg/kg) and fluoxetine (20mg/kg) were pretreated intragastrically once daily for 7 consecutive days. LPS (0.5mg/kg) was injected subcutaneously to establish the depression model 30min after pretreatment on day 7. Interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels in serum and hippocampus were detected by enzyme-linked immunosorbent assay (ELISA). Behavioral assessment was conduct 24h post LPS injection. The expressions of p65NF-κB, IκBα, brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase B (TrkB) in hippocampus were determined by western blot. The obtained results showed that liquiritigenin effectively reduced the levels of pro-inflammatory cytokines and the expressions of p-p65NF-κB and p-IκBα. Furthermore, liquiritigenin preconditioning could down-regulate the immobility time in tail suspension test (TST), forced swimming test (FST) and up-regulate BDNF and TrkB contents in hippocampus. Thus, it is assumed that the antidepressant activity of liquiritigenin might be attributed to its anti-inflammatory property and BDNF/TrkB signaling pathway.
Collapse
|
32
|
Gavin DP, Kusumo H, Sharma RP, Guizzetti M, Guidotti A, Pandey SC. Gadd45b and N-methyl-D-aspartate induced DNA demethylation in postmitotic neurons. Epigenomics 2016; 7:567-79. [PMID: 26111030 DOI: 10.2217/epi.15.12] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM In nondividing neurons examine the role of Gadd45b in active 5-methylcytosine (5MC) and 5-hydroxymethylcytosine (5HMC) removal at a gene promoter highly implicated in mental illnesses and cognition, Bdnf. MATERIALS & METHODS Mouse primary cortical neuronal cultures with and without Gadd45b siRNA transfection were treated with N-methyl-d-aspartate (NMDA). Expression changes of genes reportedly involved in DNA demethylation, Bdnf mRNA and protein and 5MC and 5HMC at Bdnf promoters were measured. RESULTS Gadd45b siRNA transfection in neurons abolishes the NMDA-induced increase in Bdnf IXa mRNA and reductions in 5MC and 5HMC at the Bdnf IXa promoter. CONCLUSION These results contribute to our understanding of DNA demethylation mechanisms in neurons, and its role in regulating NMDA responsive genes implicated in mental illnesses.
Collapse
Affiliation(s)
- David P Gavin
- Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue (M/C 151), Chicago, IL 60612, USA.,Department of Psychiatry, University of Illinois at Chicago, 1601 W Taylor St, Chicago, IL 60612, USA
| | - Handojo Kusumo
- Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue (M/C 151), Chicago, IL 60612, USA.,Department of Psychiatry, University of Illinois at Chicago, 1601 W Taylor St, Chicago, IL 60612, USA
| | - Rajiv P Sharma
- Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue (M/C 151), Chicago, IL 60612, USA.,Department of Psychiatry, University of Illinois at Chicago, 1601 W Taylor St, Chicago, IL 60612, USA
| | - Marina Guizzetti
- Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue (M/C 151), Chicago, IL 60612, USA.,Department of Psychiatry, University of Illinois at Chicago, 1601 W Taylor St, Chicago, IL 60612, USA.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR and VA Portland Health Care System, Portland, OR
| | - Alessandro Guidotti
- Department of Psychiatry, University of Illinois at Chicago, 1601 W Taylor St, Chicago, IL 60612, USA
| | - Subhash C Pandey
- Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue (M/C 151), Chicago, IL 60612, USA.,Department of Psychiatry, University of Illinois at Chicago, 1601 W Taylor St, Chicago, IL 60612, USA
| |
Collapse
|
33
|
Abstract
UNLABELLED Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, regulates both survival and differentiation of several neuronal populations in the nervous system during development, as well as synaptic plasticity in the adult brain. BDNF exerts its biological functions through its receptor TrkB. Although the regulation of BDNF transcription by neuronal activity has been widely studied, little is known about TrkB signaling-dependent expression of BDNF. Using rat primary cortical neuron cultures, we show that the BDNF gene is a subject to an extensive autoregulatory loop, where TrkB signaling upregulates the expression of all major BDNF transcripts, mainly through activating MAPK pathways. Investigating the mechanisms behind this autoregulation, we found that AP-1 transcription factors, comprising Jun and Fos family members, participate in the induction of BDNF exon I, III, and VI transcripts. AP-1 transcription factors directly upregulate the expression of exon I transcripts by binding two novel AP-1 cis-elements in promoter I. Moreover, our results show that the effect of AP-1 proteins on the activity of rat BDNF promoters III and VI is indirect, because AP-1 proteins were not detected to bind the respective promoter regions by chromatin immunoprecipitation (ChIP). Collectively, we describe an extensive positive feedback system in BDNF regulation, adding a new layer to the elaborate control of BDNF gene expression. SIGNIFICANCE STATEMENT Here, we show for the first time that in rat primary cortical neurons the expression of all major BDNF transcripts (exon I, II, III, IV, VI, and IXa transcripts) is upregulated in response to TrkB signaling, and that AP-1 transcription factors participate in the induction of exon I, III, and VI transcripts. Moreover, we have described two novel functional AP-1 cis-elements in BDNF promoter I, responsible for the activation of the promoter in response to TrkB signaling. Our results indicate the existence of a positive feedback loop for obtaining sufficient BDNF levels necessary for various TrkB signaling-dependent physiological outcomes in neurons.
Collapse
|
34
|
Cho CH, Kim J, Ahn JY, Hahn HG, Cho SW. N-adamantyl-4-methylthiazol-2-amine suppresses lipopolysaccharide-induced brain inflammation by regulating NF-κB signaling in mice. J Neuroimmunol 2015; 289:98-104. [PMID: 26616878 DOI: 10.1016/j.jneuroim.2015.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/20/2015] [Accepted: 10/24/2015] [Indexed: 01/13/2023]
Abstract
We report that N-adamantyl-4-methylthiazol-2-amine (KHG26693), a novel thiazole derivative, can prevent lipopolysaccharide (LPS)-induced brain inflammation in mice. In this LPS-induced model of brain inflammation, administration of KHG26693 effectively prevented increases in the levels of IL-1β, TNF-α, prostaglandin E2, malondialdehyde, and nitric oxide, and mitigated reductions in the levels of superoxide dismutase in the hippocampus. KHG26693 also prevented reductions in the levels of hippocampal brain-derived neurotrophic factors. Furthermore, pretreatment with KHG26693 prior to LPS treatment dramatically attenuated the elevation of inducible nitric oxide synthase and cyclooxygenase-2 protein levels. Moreover, pretreatment with KHG26693 significantly suppressed LPS-induced phosphorylation of NF-κB and IκBα through the inactivation of IKKβ. Additionally, KHG26693 caused the downregulation of LPS-induced cystathionine-b-synthase gene expression in the brain. Although the clinical relevance of our findings remains to be determined, our data suggest that KHG26693 might prevent neuronal cell injury via the reduction of inflammation and oxidative stress in the brain.
Collapse
Affiliation(s)
- Chang Hun Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Jiae Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Republic of Korea
| | - Hoh-Gyu Hahn
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul 136-791, Republic of Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea.
| |
Collapse
|
35
|
Fefelova EV, Tereshkov PP, Dutov AA, Tsybikov NN. Lymphocyte Subpopulations and Cytokine Levels in Experimental Hyperhomocysteinemia. Bull Exp Biol Med 2015. [PMID: 26212809 DOI: 10.1007/s10517-015-2962-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Moderate exogenous hyperhomocysteinemia is associated with an increase in lymphocyte count (mainly at the expense of cytotoxic T cells), a 2-fold drop of the immunoregulatory index, and increase in TNF-α, IFN-γ, and IL-17α concentrations in the serum and in mononuclear cells. Homocysteine concentration in the mononuclears remains unchanged.
Collapse
|
36
|
Mao XY, Cao YG, Ji Z, Zhou HH, Liu ZQ, Sun HL. Topiramate protects against glutamate excitotoxicity via activating BDNF/TrkB-dependent ERK pathway in rodent hippocampal neurons. Prog Neuropsychopharmacol Biol Psychiatry 2015; 60:11-7. [PMID: 25661849 DOI: 10.1016/j.pnpbp.2015.01.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/15/2015] [Accepted: 01/29/2015] [Indexed: 10/24/2022]
Abstract
Topiramate (TPM) was previously found to have neuroprotection against neuronal injury in epileptic and ischemic models. However, whether TPM protects against glutamate-induced excitotoxicity in hippocampal neurons is elusive. Our present work aimed to evaluate the protective effect of TPM against glutamate toxicity in hippocampal neurons and further figure out the potential molecular mechanisms. The in vitro glutamate excitotoxic model was prepared with 125μM glutamate for 20min. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) analysis and Hoechst 33342 staining were conducted to detect neuronal survival. The protein expressions of brain-derived neurotrophic factor (BDNF), TrkB, mitogen-activated protein kinase (MAPK) cascade (including extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 MAPK), cyclic AMP response element binding protein (CREB), Bcl-2, Bax and β-actin were detected via Western blot assay. Our results demonstrated that TPM protected hippocampal neurons from glutamate toxicity. Meanwhile, the pretreatment of TPM for 10min significantly prevented the down-regulation of BDNF and the phosphorylation of TrkB. Furthermore, the elevation of phosphorylated EKR expression was significantly inhibited after blockade of TrkB by TrkB IgG, while no alterations of phosphorylated JNK and p38 MAPK were found in the cultured hippocampal neurons. Besides, it was also found that the enhanced phosphorylation of CREB was evidently reversed under excitotoxic conditions after treating with U0126 (the selective inhibitor of ERK). The protein level of Bcl-2 was also observed to be remarkably increased after TPM treatment. In conclusion, these findings implicate that TPM exerts neuroprotective effects against glutamate excitotoxicity in hippocampal neurons and its protection may be modulated through BDNF/TrkB-dependent ERK pathway.
Collapse
Affiliation(s)
- Xiao-Yuan Mao
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, China
| | - Yong-Gang Cao
- Department of Pharmacology, Daqing Campus of Harbin Medical University, Daqing 163319, China.
| | - Zhong Ji
- Department of Physiology, Daqing Campus of Harbin Medical University, Daqing 163319, China
| | - Hong-Hao Zhou
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, China
| | - Zhao-Qian Liu
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, China.
| | - Hong-Li Sun
- Department of Pharmacology, Daqing Campus of Harbin Medical University, Daqing 163319, China.
| |
Collapse
|
37
|
Lee JH, Zhang J, Wei L, Yu SP. Neurodevelopmental implications of the general anesthesia in neonate and infants. Exp Neurol 2015; 272:50-60. [PMID: 25862287 DOI: 10.1016/j.expneurol.2015.03.028] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
Each year, about six million children, including 1.5 million infants, in the United States undergo surgery with general anesthesia, often requiring repeated exposures. However, a crucial question remains of whether neonatal anesthetics are safe for the developing central nervous system (CNS). General anesthesia encompasses the administration of agents that induce analgesic, sedative, and muscle relaxant effects. Although the mechanisms of action of general anesthetics are still not completely understood, recent data have suggested that anesthetics primarily modulate two major neurotransmitter receptor groups, either by inhibiting N-methyl-D-aspartate (NMDA) receptors, or conversely by activating γ-aminobutyric acid (GABA) receptors. Both of these mechanisms result in the same effect of inhibiting excitatory activity of neurons. In developing brains, which are more sensitive to disruptions in activity-dependent plasticity, this transient inhibition may have longterm neurodevelopmental consequences. Accumulating reports from preclinical studies show that anesthetics in neonates cause cellular toxicity including apoptosis and neurodegeneration in the developing brain. Importantly, animal and clinical studies indicate that exposure to general anesthetics may affect CNS development, resulting in long-lasting cognitive and behavioral deficiencies, such as learning and memory deficits, as well as abnormalities in social memory and social activity. While the casual relationship between cellular toxicity and neurological impairments is still not clear, recent reports in animal experiments showed that anesthetics in neonates can affect neurogenesis, which could be a possible mechanism underlying the chronic effect of anesthetics. Understanding the cellular and molecular mechanisms of anesthetic effects will help to define the scope of the problem in humans and may lead to preventive and therapeutic strategies. Therefore, in this review, we summarize the current evidence on neonatal anesthetic effects in the developmental CNS and discuss how factors influencing these processes can be translated into new therapeutic strategies.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James Zhang
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for Visual and Neurocognitive Rehabilitation, VA Medical Center, Atlanta, GA 30033, USA.
| |
Collapse
|
38
|
Baranova KA, Rybnikova EA, Samoilov MO. The neurotrophin BDNF is involved in the development and prevention of stress-induced psychopathologies. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415020038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
39
|
Alpha-linolenic acid: an omega-3 fatty acid with neuroprotective properties-ready for use in the stroke clinic? BIOMED RESEARCH INTERNATIONAL 2015; 2015:519830. [PMID: 25789320 PMCID: PMC4350958 DOI: 10.1155/2015/519830] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 09/08/2014] [Indexed: 01/29/2023]
Abstract
Alpha-linolenic acid (ALA) is plant-based essential omega-3 polyunsaturated fatty acids that must be obtained through the diet. This could explain in part why the severe deficiency in omega-3 intake pointed by numerous epidemiologic studies may increase the brain's vulnerability representing an important risk factor in the development and/or deterioration of certain cardio- and neuropathologies. The roles of ALA in neurological disorders remain unclear, especially in stroke that is a leading cause of death. We and others have identified ALA as a potential nutraceutical to protect the brain from stroke, characterized by its pleiotropic effects in neuroprotection, vasodilation of brain arteries, and neuroplasticity. This review highlights how chronic administration of ALA protects against rodent models of hypoxic-ischemic injury and exerts an anti-depressant-like activity, effects that likely involve multiple mechanisms in brain, and may be applied in stroke prevention. One major effect may be through an increase in mature brain-derived neurotrophic factor (BDNF), a widely expressed protein in brain that plays critical roles in neuronal maintenance, and learning and memory. Understanding the precise roles of ALA in neurological disorders will provide the underpinnings for the development of new therapies for patients and families who could be devastated by these disorders.
Collapse
|
40
|
Lauro C, Catalano M, Di Paolo E, Chece G, de Costanzo I, Trettel F, Limatola C. Fractalkine/CX3CL1 engages different neuroprotective responses upon selective glutamate receptor overactivation. Front Cell Neurosci 2015; 8:472. [PMID: 25653593 PMCID: PMC4301004 DOI: 10.3389/fncel.2014.00472] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/30/2014] [Indexed: 11/16/2022] Open
Abstract
Neuronal death induced by overactivation of N-methyl-d-aspartate receptors (NMDARs) is implicated in the pathophysiology of many neurodegenerative diseases such as stroke, epilepsy and traumatic brain injury. This toxic effect is mainly mediated by NR2B-containing extrasynaptic NMDARs, while NR2A-containing synaptic NMDARs contribute to cell survival, suggesting the possibility of therapeutic approaches targeting specific receptor subunits. We report that fractalkine/CX3CL1 protects hippocampal neurons from NMDA-induced cell death with a mechanism requiring the adenosine receptors type 2A (A2AR). This is different from CX3CL1-induced protection from glutamate (Glu)-induced cell death, that fully depends on A1R and requires in part A3R. We show that CX3CL1 neuroprotection against NMDA excitotoxicity involves D-serine, a co-agonist of NR2A/NMDAR, resulting in cyclic AMP-dependent transcription factor cyclic-AMP response element-binding protein (CREB) phosphorylation.
Collapse
Affiliation(s)
- Clotilde Lauro
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome Rome, Italy ; Istituto di Ricovero e Cura a Carattere Scientifico NeuroMed Pozzilli, Italy
| | | | - Giuseppina Chece
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome Rome, Italy
| | - Ida de Costanzo
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome Rome, Italy
| | - Flavia Trettel
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza University of Rome Rome, Italy ; Istituto di Ricovero e Cura a Carattere Scientifico NeuroMed Pozzilli, Italy
| |
Collapse
|
41
|
Leandra C, Tasca CI, Boeck CR. The Role of NMDA Receptors in the Development of Brain Resistance through Pre- and Postconditioning. Aging Dis 2014; 5:430-41. [PMID: 25489494 DOI: 10.14336/ad.2014.0500430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/30/2014] [Accepted: 02/09/2014] [Indexed: 11/01/2022] Open
Abstract
Brain tolerance or resistance can be achieved by interventions before and after injury through potential toxic agents used in low stimulus or dose. For brain diseases, the neuroprotection paradigm desires an attenuation of the resulting motor, cognitive, emotional, or memory deficits following the insult. Preconditioning is a well-established experimental and clinical translational strategy with great beneficial effects, but limited applications. NMDA receptors have been reported as protagonists in the adjacent cellular mechanisms contributing to the development of brain tolerance. Postconditioning has recently emerged as a new neuroprotective strategy, which has shown interesting results when applied immediately, i.e. several hours to days, after a stroke event. Investigations using chemical postconditioning are still incipient, but nevertheless represent an interesting and promising clinical strategy. In the present review pre- and postconditioning are discussed as neuroprotective paradigms and the focus of our attention lies on the participation of NMDA receptors proteins in the processes related to neuroprotection.
Collapse
Affiliation(s)
| | - Carla Inês Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Carina Rodrigues Boeck
- Laboratório de Biologia Celular e Molecular, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-graduação Ciências da Saúde, Universidade do Extremo Sul Catarinense-UNESC, Criciúma, 88806-000, SC, Brazil
| |
Collapse
|
42
|
NMDA receptor dysregulation in chronic state: A possible mechanism underlying depression with BDNF downregulation. Neurochem Int 2014; 79:88-97. [DOI: 10.1016/j.neuint.2014.09.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 11/23/2022]
|
43
|
Location- and Subunit-Specific NMDA Receptors Determine the Developmental Sevoflurane Neurotoxicity Through ERK1/2 Signaling. Mol Neurobiol 2014; 53:216-230. [DOI: 10.1007/s12035-014-9005-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
|
44
|
Vasconcelos AR, Yshii LM, Viel TA, Buck HS, Mattson MP, Scavone C, Kawamoto EM. Intermittent fasting attenuates lipopolysaccharide-induced neuroinflammation and memory impairment. J Neuroinflammation 2014; 11:85. [PMID: 24886300 PMCID: PMC4041059 DOI: 10.1186/1742-2094-11-85] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 04/21/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Systemic bacterial infections often result in enduring cognitive impairment and are a risk factor for dementia. There are currently no effective treatments for infection-induced cognitive impairment. Previous studies have shown that intermittent fasting (IF) can increase the resistance of neurons to injury and disease by stimulating adaptive cellular stress responses. However, the impact of IF on the cognitive sequelae of systemic and brain inflammation is unknown. METHODS Rats on IF for 30 days received 1 mg/kg of lipopolysaccharide (LPS) or saline intravenously. Half of the rats were subjected to behavioral tests and the other half were euthanized two hours after LPS administration and the hippocampus was dissected and frozen for analyses. RESULTS Here, we report that IF ameliorates cognitive deficits in a rat model of sepsis by a mechanism involving NF-κB activation, suppression of the expression of pro-inflammatory cytokines, and enhancement of neurotrophic support. Treatment of rats with LPS resulted in deficits in cognitive performance in the Barnes maze and inhibitory avoidance tests, without changing locomotor activity, that were ameliorated in rats that had been maintained on the IF diet. IF also resulted in reduced levels of mRNAs encoding the LPS receptor TLR4 and inducible nitric oxide synthase (iNOS) in the hippocampus. Moreover, IF prevented LPS-induced elevation of IL-1α, IL-1β and TNF-α levels, and prevented the LPS-induced reduction of BDNF levels in the hippocampus. IF also significantly attenuated LPS-induced elevations of serum IL-1β, IFN-γ, RANTES, TNF-α and IL-6 levels. CONCLUSIONS Taken together, our results suggest that IF induces adaptive responses in the brain and periphery that can suppress inflammation and preserve cognitive function in an animal model of systemic bacterial infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-900, Brazil.
| | | |
Collapse
|
45
|
Stetler RA, Leak RK, Gan Y, Li P, Zhang F, Hu X, Jing Z, Chen J, Zigmond MJ, Gao Y. Preconditioning provides neuroprotection in models of CNS disease: paradigms and clinical significance. Prog Neurobiol 2014; 114:58-83. [PMID: 24389580 PMCID: PMC3937258 DOI: 10.1016/j.pneurobio.2013.11.005] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/14/2022]
Abstract
Preconditioning is a phenomenon in which brief episodes of a sublethal insult induce robust protection against subsequent lethal injuries. Preconditioning has been observed in multiple organisms and can occur in the brain as well as other tissues. Extensive animal studies suggest that the brain can be preconditioned to resist acute injuries, such as ischemic stroke, neonatal hypoxia/ischemia, surgical brain injury, trauma, and agents that are used in models of neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. Effective preconditioning stimuli are numerous and diverse, ranging from transient ischemia, hypoxia, hyperbaric oxygen, hypothermia and hyperthermia, to exposure to neurotoxins and pharmacological agents. The phenomenon of "cross-tolerance," in which a sublethal stress protects against a different type of injury, suggests that different preconditioning stimuli may confer protection against a wide range of injuries. Research conducted over the past few decades indicates that brain preconditioning is complex, involving multiple effectors such as metabolic inhibition, activation of extra- and intracellular defense mechanisms, a shift in the neuronal excitatory/inhibitory balance, and reduction in inflammatory sequelae. An improved understanding of brain preconditioning should help us identify innovative therapeutic strategies that prevent or at least reduce neuronal damage in susceptible patients. In this review, we focus on the experimental evidence of preconditioning in the brain and systematically survey the models used to develop paradigms for neuroprotection, and then discuss the clinical potential of brain preconditioning.
Collapse
Affiliation(s)
- R Anne Stetler
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Yu Gan
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Peiying Li
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Feng Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoming Hu
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Zheng Jing
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Michael J Zigmond
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China.
| |
Collapse
|
46
|
Expression of full-length and truncated trkB in human striatum and substantia nigra neurons: implications for Parkinson's disease. J Mol Histol 2013; 45:349-61. [PMID: 24374887 DOI: 10.1007/s10735-013-9562-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/17/2013] [Indexed: 12/20/2022]
Abstract
Brain derived neurotrophic factor (BDNF) is a potent mediator of cell survival and differentiation and can reverse neuronal injury associated with Parkinson's disease (PD). Tropomyosin receptor kinase B (trkB) is the high affinity receptor for BDNF. There are two major trkB isoforms, the full-length receptor (trkB.tk(+)) and the truncated receptor (trkB.t1), that mediate the diverse, region specific functions of BDNF. Both trkB isoforms are widely distributed throughout the brain, but the isoform specific distribution of trkB.t1 and trkB.tk(+) to human neurons is not well characterized. Therefore, we report the regional and neuronal distribution of trkB.tk(+) and trkB.t1 in the striatum and substantia nigra pars compacta (SNpc) of human autopsy tissues from control and PD cases. In both PD and control tissues, we found abundant, punctate distribution of trkB.tk(+) and trkB.t1 proteins in striatum and SNpc neurons. In PD, trkB.tk(+) is decreased in striatal neurites, increased in striatal somata, decreased in SNpc somata and dendrites, and increased in SNpc axons. TrkB.t1 is increased in striatal somata, decreased in striatal axons, and increased in SNpc distal dendrites. We believe changes in trkB isoform distribution and expression levels may be markers of pathology and affect the neuronal response to BDNF.
Collapse
|
47
|
Modulation of NMDAR subunit expression by TRPM2 channels regulates neuronal vulnerability to ischemic cell death. J Neurosci 2013; 33:17264-77. [PMID: 24174660 DOI: 10.1523/jneurosci.1729-13.2013] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal vulnerability to ischemia is dependent on the balance between prosurvival and prodeath cellular signaling. In the latter, it is increasingly appreciated that toxic Ca(2+) influx can occur not only via postsynaptic glutamate receptors, but also through other cation conductances. One such conductance, the Transient receptor potential melastatin type-2 (TRPM2) channel, is a nonspecific cation channel having homology to TRPM7, a conductance reported to play a key role in anoxic neuronal death. The role of TRPM2 conductances in ischemic Ca(2+) influx has been difficult to study because of the lack of specific modulators. Here we used TRPM2-null mice (TRPM2(-/-)) to study how TRPM2 may modulate neuronal vulnerability to ischemia. TRPM2(-/-) mice subjected to transient middle cerebral artery occlusion exhibited smaller infarcts when compared with wild-type animals, suggesting that the absence of TRPM2 is neuroprotective. Surprisingly, field potentials (fEPSPs) recorded during redox modulation in brain slices taken from TRPM2(-/-) mice revealed increased excitability, a phenomenon normally associated with ischemic vulnerability, whereas wild-type fEPSPs were unaffected. The upregulation in fEPSP in TRPM2(-/-) neurons was blocked selectively by a GluN2A antagonist. This increase in excitability of TRPM2(-/-) fEPSPs during redox modulation depended on the upregulation and downregulation of GluN2A- and GluN2B-containing NMDARs, respectively, and on augmented prosurvival signaling via Akt and ERK pathways culminating in the inhibition of the proapoptotic factor GSK3β. Our results suggest that TRPM2 plays a role in downregulating prosurvival signals in central neurons and that TRPM2 channels may comprise a therapeutic target for preventing ischemic damage.
Collapse
|
48
|
Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol 2013; 115:157-88. [PMID: 24361499 DOI: 10.1016/j.pneurobio.2013.11.006] [Citation(s) in RCA: 819] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/28/2013] [Accepted: 11/29/2013] [Indexed: 01/22/2023]
Abstract
Excitotoxicity, the specific type of neurotoxicity mediated by glutamate, may be the missing link between ischemia and neuronal death, and intervening the mechanistic steps that lead to excitotoxicity can prevent stroke damage. Interest in excitotoxicity began fifty years ago when monosodium glutamate was found to be neurotoxic. Evidence soon demonstrated that glutamate is not only the primary excitatory neurotransmitter in the adult brain, but also a critical transmitter for signaling neurons to degenerate following stroke. The finding led to a number of clinical trials that tested inhibitors of excitotoxicity in stroke patients. Glutamate exerts its function in large by activating the calcium-permeable ionotropic NMDA receptor (NMDAR), and different subpopulations of the NMDAR may generate different functional outputs, depending on the signaling proteins directly bound or indirectly coupled to its large cytoplasmic tail. Synaptic activity activates the GluN2A subunit-containing NMDAR, leading to activation of the pro-survival signaling proteins Akt, ERK, and CREB. During a brief episode of ischemia, the extracellular glutamate concentration rises abruptly, and stimulation of the GluN2B-containing NMDAR in the extrasynaptic sites triggers excitotoxic neuronal death via PTEN, cdk5, and DAPK1, which are directly bound to the NMDAR, nNOS, which is indirectly coupled to the NMDAR via PSD95, and calpain, p25, STEP, p38, JNK, and SREBP1, which are further downstream. This review aims to provide a comprehensive summary of the literature on excitotoxicity and our perspectives on how the new generation of excitotoxicity inhibitors may succeed despite the failure of the previous generation of drugs.
Collapse
Affiliation(s)
- Ted Weita Lai
- Graduate Institute of Clinical Medical Science, China Medical University, 91 Hsueh-Shih Road, 40402 Taichung, Taiwan; Translational Medicine Research Center, China Medical University Hospital, 2 Yu-De Road, 40447 Taichung, Taiwan.
| | - Shu Zhang
- Translational Medicine Research Center, China Medical University Hospital, 2 Yu-De Road, 40447 Taichung, Taiwan; Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, V6T 2B5 Vancouver, Canada
| | - Yu Tian Wang
- Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, V6T 2B5 Vancouver, Canada.
| |
Collapse
|
49
|
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]
|
50
|
Rothman SM, Mattson MP. Activity-dependent, stress-responsive BDNF signaling and the quest for optimal brain health and resilience throughout the lifespan. Neuroscience 2013; 239:228-40. [PMID: 23079624 PMCID: PMC3629379 DOI: 10.1016/j.neuroscience.2012.10.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 09/24/2012] [Accepted: 10/05/2012] [Indexed: 12/31/2022]
Abstract
During development of the nervous system, the formation of connections (synapses) between neurons is dependent upon electrical activity in those neurons, and neurotrophic factors produced by target cells play a pivotal role in such activity-dependent sculpting of the neural networks. A similar interplay between neurotransmitter and neurotrophic factor signaling pathways mediates adaptive responses of neural networks to environmental demands in adult mammals, with the excitatory neurotransmitter glutamate and brain-derived neurotrophic factor (BDNF) being particularly prominent regulators of synaptic plasticity throughout the central nervous system. Optimal brain health throughout the lifespan is promoted by intermittent challenges such as exercise, cognitive stimulation and dietary energy restriction, that subject neurons to activity-related metabolic stress. At the molecular level, such challenges to neurons result in the production of proteins involved in neurogenesis, learning and memory and neuronal survival; examples include proteins that regulate mitochondrial biogenesis, protein quality control, and resistance of cells to oxidative, metabolic and proteotoxic stress. BDNF signaling mediates up-regulation of several such proteins including the protein chaperone GRP-78, antioxidant enzymes, the cell survival protein Bcl-2, and the DNA repair enzyme APE1. Insufficient exposure to such challenges, genetic factors may conspire to impair BDNF production and/or signaling resulting in the vulnerability of the brain to injury and neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Further, BDNF signaling is negatively regulated by glucocorticoids. Glucocorticoids impair synaptic plasticity in the brain by negatively regulating spine density, neurogenesis and long-term potentiation, effects that are potentially linked to glucocorticoid regulation of BDNF. Findings suggest that BDNF signaling in specific brain regions mediates some of the beneficial effects of exercise and energy restriction on peripheral energy metabolism and the cardiovascular system. Collectively, the findings described in this article suggest the possibility of developing prescriptions for optimal brain health based on activity-dependent BDNF signaling.
Collapse
Affiliation(s)
- S M Rothman
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | | |
Collapse
|