1
|
Liu M, Shi S, Guo Y, Lv M, Feng X, Liu T, Yang H, Zhang Z. Potential Biomarkers for Neurobrucellosis Diagnosis and Treatment Based on Cerebrospinal Fluid Metabolomics Analysis. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e9946. [PMID: 39601633 DOI: 10.1002/rcm.9946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Zhenzhu Tongluo pills (ZZTL) have been utilized for the treatment of neurobrucellosis (NB) in the Inner Mongolia region of China. However, the specific mechanism underlying the neuroprotective effects of ZZTL in NB remains insufficiently explored. Therefore, this study aimed to analyze the metabolite profiles across different groups to identify potential biomarkers for the diagnosis and treatment of NB. METHODS LC-MS analysis was used to screen differential metabolites in cerebrospinal fluid (CSF) samples from control subjects, NB patients, and NB patients undergoing ZZTL therapy. RESULTS We identified 225 common metabolites across two comparison groups (NB vs. control group and NB + ZZTL vs. NB group). Among these metabolites, 138 were downregulated, and 87 were upregulated in the NB group compared to the control group, while the levels of these metabolites were reversed by ZZTL therapy. Creatinine exhibited the highest VIP score in both comparison groups, with elevated levels observed in CSF samples from NB patients compared to controls; however, its levels were reduced following ZZTL therapy. Additionally, hypoxanthine and uric acids levels were also increased in CSF samples from NB patients, indicating dysregulation of purine metabolism in NB; however, these changes were reversed by ZZTL. Furthermore, pantothenic acid (vitamin B5) and niacinamide (vitamin B3) levels were decreased in CSF samples from NB patients, suggesting a potential link between NB and vitamin B3 and B5 deficiency; however, these changes were reversed by ZZTL. CONCLUSION Collectively, CSF metabolomics may effectively differentiate NB patients from control subjects, providing valuable insights for NB diagnosis and treatment.
Collapse
Affiliation(s)
- Meiling Liu
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Shujun Shi
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yuzhou Guo
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Menghan Lv
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xuan Feng
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Ting Liu
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hao Yang
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital & Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhelin Zhang
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
2
|
Arslan R, Doganay S, Budak O, Bahtiyar N. Investigation of preconditioning and the protective effects of nicotinamide against cerebral ischemia-reperfusion injury in rats. Neurosci Lett 2024; 840:137949. [PMID: 39181500 DOI: 10.1016/j.neulet.2024.137949] [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: 02/27/2024] [Revised: 08/04/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This study investigated the antioxidant and neuroprotective effects of nicotinamide combined with ischemic preconditioning against cerebral ischemia reperfusion (CIR) injury. Thirty-five Wistar albino male rats were randomly divided into five groups: sham, preconditioned ischemia/reperfusion (IP+IR), ischemia/reperfusion (IR), preconditioned ischemia/reperfusion + nicotinamide (IP+IR+N), and ischemia/reperfusion + nicotinamide (IR+N). CIR was achieved with bilateral common carotid artery occlusion. IP+IR and IP+IR+N groups 30 min before ischemia; Three cycles of 10 sec ischemia/30 sec reperfusion followed by 20 min IR were applied. The IP+IR+N and IR+N groups received 500 mg/kg nicotinamide intraperitoneally. After 24 h of reperfusion, a neurological evaluation was performed and vertıcal pole test. Biochemically, malondialdehyde (MDA), glutathione (GSH) levels and catalase (CAT) activity were measured in blood and brain tissue samples. Rates of red neurons, sateliosis and spongiosis were determined histopathologically in the prefrontal cortex areas. After CIR, MDA levels increased significantly in serum and brain tissue in the IR group compared to the sham group, while GSH and CAT activity decreased in the brain tissue (p < 0.05). MDA levels in the tissues were found significantly decreased in the IR+N group compared to the IR group (p < 0.05). Administration of nicotinamide together with IP significantly decreased MDA levels in brain tissue and increased GSH and CAT activity (p < 0.05). Compared to the IR group, the morphological and neurological damage in the prefrontal cortex areas decreased in the IP+IR, IP+IR+N, and IR+N groups (p < 0.05). In addition, red neuron, sateliosis and spongiosis rates increased significantly in the IR group compared to the Sham, IP+IR+N, IR+N groups (p < 0.001 for all). In neurological evaluation, while the neurological score increased and the time on the vertical pole decreased significantly in the IR group, preconditioning, and nicotinamide groups reversed (p < 0.05). The study's results show that nicotinamide administration with ischemic preconditioning alleviates cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ruhat Arslan
- Istinye University, Faculty of Medicine, Department of Physiology, TR - 34000 Istanbul, Turkey; Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, TR-34098 Istanbul, Turkey.
| | - Songul Doganay
- Sakarya University, Faculty of Medicine, Department of Physiology, TR-54000 Sakarya, Turkey.
| | - Ozcan Budak
- Sakarya University, Faculty of Medicine, Department of Histology and Embryology, TR-54000 Sakarya, Turkey.
| | - Nurten Bahtiyar
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Biophysics, TR-34098 Istanbul, Turkey.
| |
Collapse
|
3
|
Joshi SM, Thomas TC, Jadavji NM. Impact of increasing one-carbon metabolites on traumatic brain injury outcome using pre-clinical models. Neural Regen Res 2024; 19:1728-1733. [PMID: 38103238 PMCID: PMC10960300 DOI: 10.4103/1673-5374.389629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/20/2023] [Accepted: 10/17/2023] [Indexed: 12/18/2023] Open
Abstract
Traumatic brain injury is a major cause of death and disability worldwide, affecting over 69 million individuals yearly. One-carbon metabolism has been shown to have beneficial effects after brain damage, such as ischemic stroke. However, whether increasing one-carbon metabolite vitamins impacts traumatic brain injury outcomes in patients requires more investigation. The aim of this review is to evaluate how one-carbon metabolites impact outcomes after the onset of traumatic brain injury. PubMed, Web of Science, and Google Scholar databases were searched for studies that examined the impact of B-vitamin supplementation on traumatic brain injury outcomes. The search terms included combinations of the following words: traumatic brain injury, dietary supplementation, one-carbon metabolism, and B-vitamins. The focus of each literature search was basic science data. The year of publication in the literature searches was not limited. Our analysis of the literature has shown that dietary supplementation of B-vitamins has significantly improved the functional and behavioral recovery of animals with traumatic brain injury compared to controls. However, this improvement is dosage-dependent and is contingent upon the onset of supplementation and whether there is a sustained or continuous delivery of vitamin supplementation post-traumatic brain injury. The details of supplementation post-traumatic brain injury need to be further investigated. Overall, we conclude that B-vitamin supplementation improves behavioral outcomes and reduces cognitive impairment post-traumatic brain injury in animal model systems. Further investigation in a clinical setting should be strongly considered in conjunction with current medical treatments for traumatic brain injury-affected individuals.
Collapse
Affiliation(s)
- Sanika M. Joshi
- College of Osteopathic Medicine, Midwestern University, Glendale, CA, USA
- Department of Biomedical Sciences, Midwestern University, Glendale, CA, USA
| | - Theresa Currier Thomas
- Department of Child Health, College of Medicine – Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Nafisa M. Jadavji
- College of Osteopathic Medicine, Midwestern University, Glendale, CA, USA
- Department of Biomedical Sciences, Midwestern University, Glendale, CA, USA
- Department of Child Health, College of Medicine – Phoenix, University of Arizona, Phoenix, AZ, USA
- College of Veterinary Medicine, Midwestern University, Glendale, CA, USA
- Department of Neuroscience, Carleton University, Ottawa, Canada
| |
Collapse
|
4
|
Denniss RJ, Barker LA. Brain Trauma and the Secondary Cascade in Humans: Review of the Potential Role of Vitamins in Reparative Processes and Functional Outcome. Behav Sci (Basel) 2023; 13:bs13050388. [PMID: 37232626 DOI: 10.3390/bs13050388] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
An estimated sixty-nine million people sustain a traumatic brain injury each year. Trauma to the brain causes the primary insult and initiates a secondary biochemical cascade as part of the immune and reparative response to injury. The secondary cascade, although a normal physiological response, may also contribute to ongoing neuroinflammation, oxidative stress and axonal injury, continuing in some cases years after the initial insult. In this review, we explain some of the biochemical mechanisms of the secondary cascade and their potential deleterious effects on healthy neurons including secondary cell death. The second part of the review focuses on the role of micronutrients to neural mechanisms and their potential reparative effects with regards to the secondary cascade after brain injury. The biochemical response to injury, hypermetabolism and excessive renal clearance of nutrients after injury increases the demand for most vitamins. Currently, most research in the area has shown positive outcomes of vitamin supplementation after brain injury, although predominantly in animal (murine) models. There is a pressing need for more research in this area with human participants because vitamin supplementation post-trauma is a potential cost-effective adjunct to other clinical and therapeutic treatments. Importantly, traumatic brain injury should be considered a lifelong process and better evaluated across the lifespan of individuals who experience brain injury.
Collapse
Affiliation(s)
- Rebecca J Denniss
- Department of Psychology, The University of Sheffield, Sheffield S10 2TN, UK
| | - Lynne A Barker
- Centre for Behavioural Science and Applied Psychology, Department of Psychology, Sociology and Politics, Sheffield Hallam University, Sheffield S1 1WB, UK
| |
Collapse
|
5
|
Jacquens A, Needham EJ, Zanier ER, Degos V, Gressens P, Menon D. Neuro-Inflammation Modulation and Post-Traumatic Brain Injury Lesions: From Bench to Bed-Side. Int J Mol Sci 2022; 23:11193. [PMID: 36232495 PMCID: PMC9570205 DOI: 10.3390/ijms231911193] [Citation(s) in RCA: 19] [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: 08/25/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Head trauma is the most common cause of disability in young adults. Known as a silent epidemic, it can cause a mosaic of symptoms, whether neurological (sensory-motor deficits), psychiatric (depressive and anxiety symptoms), or somatic (vertigo, tinnitus, phosphenes). Furthermore, cranial trauma (CT) in children presents several particularities in terms of epidemiology, mechanism, and physiopathology-notably linked to the attack of an immature organ. As in adults, head trauma in children can have lifelong repercussions and can cause social and family isolation, difficulties at school, and, later, socio-professional adversity. Improving management of the pre-hospital and rehabilitation course of these patients reduces secondary morbidity and mortality, but often not without long-term disability. One hypothesized contributor to this process is chronic neuroinflammation, which could accompany primary lesions and facilitate their development into tertiary lesions. Neuroinflammation is a complex process involving different actors such as glial cells (astrocytes, microglia, oligodendrocytes), the permeability of the blood-brain barrier, excitotoxicity, production of oxygen derivatives, cytokine release, tissue damage, and neuronal death. Several studies have investigated the effect of various treatments on the neuroinflammatory response in traumatic brain injury in vitro and in animal and human models. The aim of this review is to examine the various anti-inflammatory therapies that have been implemented.
Collapse
Affiliation(s)
- Alice Jacquens
- Unité de Neuroanesthésie-Réanimation, Hôpital de la Pitié Salpêtrière 43-87, Boulevard de l’Hôpital, F-75013 Paris, France
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - Edward J. Needham
- Division of Anaesthesia, Addenbrooke’s Hospital, University of Cambridge, Box 93, Hills Road, Cambridge CB2 2QQ, UK
| | - Elisa R. Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Vincent Degos
- Unité de Neuroanesthésie-Réanimation, Hôpital de la Pitié Salpêtrière 43-87, Boulevard de l’Hôpital, F-75013 Paris, France
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - Pierre Gressens
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - David Menon
- Division of Anaesthesia, Addenbrooke’s Hospital, University of Cambridge, Box 93, Hills Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
6
|
Macks C, Jeong D, Lee JS. Therapeutic efficacy of rolipram delivered by PgP nanocarrier on secondary injury and motor function in a rat TBI model. Nanomedicine (Lond) 2022; 17:431-445. [PMID: 35184609 PMCID: PMC8905552 DOI: 10.2217/nnm-2021-0271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop poly(lactide-co-glycolide)-graft-polyethylenimine (PgP) as a nanocarrier for the delivery of rolipram (Rm) and evaluate the therapeutic efficacy of Rm-loaded PgP (Rm-PgP) on secondary injury and motor function in a rat traumatic brain injury (TBI) model. Materials & methods: Rm-PgP was injected in the injured brain lesion immediately after TBI using a microinjection pump. Secondary injury pathologies such as inflammatory response, apoptosis and astrogliosis were assessed by histological analysis and functional recovery was assessed by assorted motor function tests. Results: Rm-PgP restored cyclic adenosine monophosphate level in the injured brain close to the sham level and Rm-PgP treatment reduced lesion volume, neuroinflammation and apoptosis and improved motor function at 7 days post-TBI. Conclusion: One single injection of Rm-PgP can be effective for acute mild TBI treatment.
Collapse
Affiliation(s)
- Christian Macks
- Department of Bioengineering, Drug Design, Development, & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Daun Jeong
- Department of Bioengineering, Drug Design, Development, & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Jeoung Soo Lee
- Department of Bioengineering, Drug Design, Development, & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
7
|
Rehman IU, Ahmad R, Khan I, Lee HJ, Park J, Ullah R, Choi MJ, Kang HY, Kim MO. Nicotinamide Ameliorates Amyloid Beta-Induced Oxidative Stress-Mediated Neuroinflammation and Neurodegeneration in Adult Mouse Brain. Biomedicines 2021; 9:biomedicines9040408. [PMID: 33920212 PMCID: PMC8070416 DOI: 10.3390/biomedicines9040408] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) is the most predominant age-related neurodegenerative disease, pathologically characterized by the accumulation of aggregates of amyloid beta Aβ1–42 and tau hyperphosphorylation in the brain. It is considered to be the primary cause of cognitive dysfunction. The aggregation of Aβ1–42 leads to neuronal inflammation and apoptosis. Since vitamins are basic dietary nutrients that organisms need for their growth, survival, and other metabolic functions, in this study, the underlying neuroprotective mechanism of nicotinamide (NAM) Vitamin B3 against Aβ1–42 -induced neurotoxicity was investigated in mouse brains. Intracerebroventricular (i.c.v.) Aβ1–42 injection elicited neuronal dysfunctions that led to memory impairment and neurodegeneration in mouse brains. After 24 h after Aβ1–42 injection, the mice were treated with NAM (250 mg/kg intraperitoneally) for 1 week. For biochemical and Western blot studies, the mice were directly sacrificed, while for confocal and “immunohistochemical staining”, mice were perfused transcardially with 4% paraformaldehyde. Our biochemical, immunofluorescence, and immunohistochemical results showed that NAM can ameliorate neuronal inflammation and apoptosis by reducing oxidative stress through lowering malondialdehyde and 2,7-dichlorofluorescein levels in an Aβ1–42-injected mouse brains, where the regulation of p-JNK further regulated inflammatory marker proteins (TNF-α, IL-1β, transcription factor NF-kB) and apoptotic marker proteins (Bax, caspase 3, PARP1). Furthermore, NAM + Aβ treatment for 1 week increased the amount of survival neurons and reduced neuronal cell death in Nissl staining. We also analyzed memory dysfunction via behavioral studies and the analysis showed that NAM could prevent Aβ1–42 -induced memory deficits. Collectively, the results of this study suggest that NAM may be a potential preventive and therapeutic candidate for Aβ1–42 -induced reactive oxygen species (ROS)-mediated neuroinflammation, neurodegeneration, and neurotoxicity in an adult mouse model.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Hyeon Jin Lee
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Jungsung Park
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Myeong Jun Choi
- Research and Development Center, Axceso Bio-pharma co, Anyang 14056, Korea;
| | - Hee Young Kang
- Department of Neurology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52828, Korea;
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
8
|
Liu B, Zhao G, Jin L, Shi J. Nicotinamide Improves Cognitive Function in Mice With Chronic Cerebral Hypoperfusion. Front Neurol 2021; 12:596641. [PMID: 33569040 PMCID: PMC7868534 DOI: 10.3389/fneur.2021.596641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/05/2021] [Indexed: 01/04/2023] Open
Abstract
Normal brain function requires steady blood supply to maintain stable energy state. When blood supply to the brain becomes suboptimal for a long period of time, chronic cerebral hypoperfusion (CCH) and a variety of brain changes may occur. CCH causes white matter injury and cognitive impairment. The present study investigated the effect of nicotinamide (NAM) on CCH-induced cognitive impairment and white matter damage in mice. Male C57Bl/6J mice aged 10–12 weeks (mean age = 11 ± 1 weeks) and weighing 24 - 29 g (mean weight = 26.5 ± 2.5 g) were randomly assigned to three groups (eight mice/group): sham group, CCH group and NAM group. Chronic cerebral hypoperfusion (CCH) was induced using standard methods. The treatment group mice received intraperitoneal injection of NAM at a dose of 200 mg/kg body weight (bwt) daily for 30 days. Learning, memory, anxiety, and depression-like behaviors were measured using Morris water maze test (MWMT), open field test (OFT), sucrose preference test (SPT), and forced swim test (FST), respectively. White matter damage and remodeling were determined via histological/ immunohistochemical analyses, and western blotting, respectively. The results showed that the time spent in target quadrant, number of crossings and escape latency were significantly lower in CCH group than in sham group, but they were significantly increased by NAM (p < 0.05). Mice in NAM group moved significantly faster and covered longer distances, when compared with those in CCH group (p < 0.05). The percentage of time spent in open arms and the number of entries to the open arms were significantly lower in CCH group than in NAM group (p < 0.05). Moreover, anhedonia and histologic scores (index of myelin injury) were significantly higher in CCH group than in sham group, but they were significantly reduced by NAM (p < 0.05). The results of immunohistochemical staining and Western blotting showed that the protein expressions of 2′, 3′-cyclic-nucleotide 3′-phosphodiesterase (CNPase) and synaptophysin were significantly downregulated in CCH group, relative to sham group, but they were significantly upregulated by NAM (p < 0.05). These results indicate that NAM improves cognitive function in mice with CCH.
Collapse
Affiliation(s)
- Bin Liu
- Department of Geriatrics, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing Brain Hospital, Nanjing, China
| | - Guifeng Zhao
- Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Ling Jin
- Department of Geriatrics, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing Brain Hospital, Nanjing, China
| | - Jingping Shi
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing Brain Hospital, Nanjing, China
| |
Collapse
|
9
|
Kang BS, Choi BY, Kho AR, Lee SH, Hong DK, Jeong JH, Kang DH, Park MK, Suh SW. An Inhibitor of the Sodium-Hydrogen Exchanger-1 (NHE-1), Amiloride, Reduced Zinc Accumulation and Hippocampal Neuronal Death after Ischemia. Int J Mol Sci 2020; 21:ijms21124232. [PMID: 32545865 PMCID: PMC7352629 DOI: 10.3390/ijms21124232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Acidosis in the brain plays an important role in neuronal injury and is a common feature of several neurological diseases. It has been reported that the sodium–hydrogen exchanger-1 (NHE-1) is a key mediator of acidosis-induced neuronal injury. It modulates the concentration of intra- and extra-cellular sodium and hydrogen ions. During the ischemic state, excessive sodium ions enter neurons and inappropriately activate the sodium–calcium exchanger (NCX). Zinc can also enter neurons through voltage-gated calcium channels and NCX. Here, we tested the hypothesis that zinc enters the intracellular space through NCX and the subsequent zinc accumulation induces neuronal cell death after global cerebral ischemia (GCI). Thus, we conducted the present study to confirm whether inhibition of NHE-1 by amiloride attenuates zinc accumulation and subsequent hippocampus neuronal death following GCI. Mice were subjected to GCI by bilateral common carotid artery (BCCA) occlusion for 30 min, followed by restoration of blood flow and resuscitation. Amiloride (10 mg/kg, intraperitoneally (i.p.)) was immediately injected, which reduced zinc accumulation and neuronal death after GCI. Therefore, the present study demonstrates that amiloride attenuates GCI-induced neuronal injury, likely via the prevention of intracellular zinc accumulation. Consequently, we suggest that amiloride may have a high therapeutic potential for the prevention of GCI-induced neuronal death.
Collapse
Affiliation(s)
- Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - A Ra Kho
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Jeong Hyun Jeong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Dong Hyeon Kang
- Department of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Korea;
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
- Correspondence: ; Tel.: +82-10-8573-6364
| |
Collapse
|
10
|
Kálmán M, Oszwald E, Pócsai K. Three-plane description of astroglial populations of OVLT subdivisions in rat: Tanycyte connections to distant parts of third ventricle. J Comp Neurol 2019; 527:2793-2812. [PMID: 31045238 DOI: 10.1002/cne.24707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 11/07/2022]
Abstract
This study demonstrates glial and gliovascular markers of organon vasculosum laminae terminalis (OVLT) in three planes. The distribution of glial markers displayed similarities to the subfornical organ. There was an inner part with vimentin- and nestin-immunopositive glia whereas GFAP and the water-channel aquaporin 4 were found at the periphery. This separation indicates different functions of the two regions. The presence of nestin may indicate stem cell-capabilities whereas aquaporin 4 has been reported to promote the osmoreceptor function. Glutamine synthetase immunoreactivity was sparse like in the area postrema and subfornical organ. The laminin and β-dystroglycan immunolabelings altered along the vessels such as in the subfornical organ indicating altering gliovascular relations. The different subdivisions of OVLT received glial processes of different origins. The posterior periventricular zone contained short vimentin-immunopositive processes from the ependyma of the adjacent surface of the third ventricle. The lateral periventricular zone received forceps-like process systems from the anterolateral part of the third ventricle. Most interestingly, the "dorsal cap" received a mixed group of long GFAP- and vimentin-immunopositive processes from a distant part of the third ventricle. The processes may have two functions: a guidance for newly produced cells like radial glia in immature brain and/or a connection between distant parts of the third ventricle and OVLT.
Collapse
Affiliation(s)
- Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Oszwald
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Károly Pócsai
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
11
|
Reetz K, Hilgers RD, Isfort S, Dohmen M, Didszun C, Fedosov K, Kistermann J, Mariotti C, Durr A, Boesch S, Klopstock T, Rodríguez de Rivera Garrido FJ, Schöls L, Klockgether T, Pandolfo M, Korinthenberg R, Lavin P, Molenberghs G, Libri V, Giunti P, Festenstein R, Schulz JB. Protocol of a randomized, double-blind, placebo-controlled, parallel-group, multicentre study of the efficacy and safety of nicotinamide in patients with Friedreich ataxia (NICOFA). Neurol Res Pract 2019; 1:33. [PMID: 33324899 PMCID: PMC7650055 DOI: 10.1186/s42466-019-0038-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 11/12/2022] Open
Abstract
Introduction Currently, no treatment that delays with the progression of Friedreich ataxia is available. In the majority of patients Friedreich ataxia is caused by homozygous pathological expansion of GAA repeats in the first intron of the FXN gene. Nicotinamide acts as a histone deacetylase inhibitor. Dose escalation studies have shown, that short term treatment with dosages of up to 4 g/day increase the expression of FXN mRNA and frataxin protein up to the levels of asymptomatic heterozygous gene carriers. The long-term effects and the effects on clinical endpoints, activities of daily living and quality of life are unknown. Methods The aim of the NICOFA study is to investigate the efficacy and safety of nicotinamide for the treatment of Friedreich ataxia over 24 months. An open-label dose adjustment wash-in period with nicotinamide (phase A: weeks 1–4) to the individually highest tolerated dose of 2–4 g nicotinamide/day will be followed by a 2 (nicotinamide group): 1 (placebo group) randomization (phase B: weeks 5–104). In the nicotinamide group, patients will continue with their individually highest tolerated dose between 2 and 4 g/d per os once daily and the placebo group patients will be receiving matching placebo. Safety assessments will consist of monitoring and recording of all adverse events and serious adverse events, regular monitoring of haematology, blood chemistry and urine values, regular measurement of vital signs and the performance of physical examinations including cardiological signs. The primary outcome is the change in the Scale for the Assessment and Rating of Ataxia (SARA) over time as compared with placebo in patients with Friedreich ataxia based on the linear mixed effect model (LMEM) model. Secondary endpoints are measures of quality of life, functional motor and cognitive measures, clinician’s and patient’s global impression-change scales as well as the up-regulation of the frataxin protein level, safety and survival/death. Perspective The NICOFA study represents one of the first attempts to assess the clinical efficacy of an epigenetic therapeutic intervention for this disease and will provide evidence of possible disease modifying effects of nicotinamide treatment in patients with Friedreich ataxia. Trial registration EudraCT-No.: 2017-002163-17, ClinicalTrials.govNCT03761511.
Collapse
Affiliation(s)
- Kathrin Reetz
- Department of Neurology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, 52074 Aachen, Germany
| | - Ralf-Dieter Hilgers
- Department of Medical Statistics, RWTH Aachen University, Pauwelsstraße 19, Aachen, Germany
| | - Susanne Isfort
- Center for Translational & Clinical Research Aachen (CTC-A), RWTH Aachen University, Pauwelsstraße 30, Aachen, Germany
| | - Marc Dohmen
- Center for Translational & Clinical Research Aachen (CTC-A), RWTH Aachen University, Pauwelsstraße 30, Aachen, Germany
| | - Claire Didszun
- Department of Neurology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, 52074 Aachen, Germany
| | - Kathrin Fedosov
- Department of Neurology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, 52074 Aachen, Germany
| | - Jennifer Kistermann
- Center for Translational & Clinical Research Aachen (CTC-A), RWTH Aachen University, Pauwelsstraße 30, Aachen, Germany
| | - Caterina Mariotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alexandra Durr
- Genetic Department, ICM (Brain and Spine Institute) Sorbonne Universités, UPMC University Paris 06 UMR S 1127, and INSERM U 1127, CNRS UMR 7225 and APHP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Sylvia Boesch
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Klopstock
- Department of Neurology, Friedrich Baur Institute, University Hospital of the Ludwig-Maximilians-Universität Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Ludger Schöls
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Thomas Klockgether
- Department of Neurology, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Massimo Pandolfo
- Laboratory of Experimental Neurology, Université Libre de Bruxelles, Brussels, Belgium
| | - Rudolf Korinthenberg
- Ethical Commission, Albert-Ludwigs-University Freiburg, Engelbergstr. 21, 79106 Freiburg, Germany
| | - Philip Lavin
- Boston Biostatistics Research Foundation, Framingham, MA USA
| | - Geert Molenberghs
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, UHasselt and KU Leuven, Leuven, Belgium
| | - Vincenzo Libri
- NIHR UCLH Clinical Research Facility-Leonard Wolfson Experimental Neurology Centre, University College London (UCL) Institute of Neurology, London, UK
| | - Paola Giunti
- Department of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, UK
| | - Richard Festenstein
- Gene Control Mechanisms and Disease Group, Department of Medicine, Division of Brain Sciences and MRC London Institute for Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Jörg B Schulz
- Department of Neurology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, 52074 Aachen, Germany.,Center for Translational & Clinical Research Aachen (CTC-A), RWTH Aachen University, Pauwelsstraße 30, Aachen, Germany
| | | |
Collapse
|
12
|
Abstract
It is increasingly recognized that tissue-specific nutrient deficiencies can exist in the absence of whole-body deficiency and that these deficiencies may result from disease or disease-related physiological processes. Brain and central nervous system tissues require adequate nutrient levels to function. Many nutrients are concentrated in the cerebrospinal fluid relative to the serum in healthy individuals, and other nutrients resist depletion in the presence of whole-body nutrient depletion. The endothelial, epithelial, and arachnoid brain barriers work in concert to selectively transport, concentrate, and maintain levels of the specific nutrients required by the brain while also blocking the passage of blood-borne toxins and pathogens to brain and central nervous system tissues. These barriers preserve nutrient levels within the brain and actively concentrate nutrients within the cerebrospinal fluid and brain. The roles of physical and energetic barriers, including the blood-brain and blood-nerve barriers, in maintaining brain nutrient levels in health and disease are discussed.
Collapse
Affiliation(s)
- Kendra A Tiani
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA;
| | - Patrick J Stover
- College of Agriculture and Life Sciences, Texas A & M University, College Station, Texas 77843-2142, USA
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA;
| |
Collapse
|
13
|
Pandya JD, Leung LY, Yang X, Flerlage WJ, Gilsdorf JS, Deng-Bryant Y, Shear DA. Comprehensive Profile of Acute Mitochondrial Dysfunction in a Preclinical Model of Severe Penetrating TBI. Front Neurol 2019; 10:605. [PMID: 31244764 PMCID: PMC6579873 DOI: 10.3389/fneur.2019.00605] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 05/22/2019] [Indexed: 01/11/2023] Open
Abstract
Mitochondria constitute a central role in brain energy metabolism, and play a pivotal role in the development of secondary pathophysiology and subsequent neuronal cell death following traumatic brain injury (TBI). Under normal circumstances, the brain consumes glucose as the preferred energy source for adenosine triphosphate (ATP) production over ketones. To understand the comprehensive picture of substrate-specific mitochondrial bioenergetics responses following TBI, adult male rats were subjected to either 10% unilateral penetrating ballistic-like brain injury (PBBI) or sham craniectomy (n = 5 animals per group). At 24 h post-injury, mitochondria were isolated from pooled brain regions (frontal cortex and striatum) of the ipsilateral hemisphere. Mitochondrial bioenergetics parameters were measured ex vivo in the presence of four sets of metabolic substrates: pyruvate+malate (PM), glutamate+malate (GM), succinate (Succ), and β-hydroxybutyrate+malate (BHBM). Additionally, mitochondrial matrix dehydrogenase activities [i.e., pyruvate dehydrogenase complex (PDHC), alpha-ketoglutarate dehydrogenase complex (α-KGDHC), and glutamate dehydrogenase (GDH)] and mitochondrial membrane-bound dehydrogenase activities [i.e., electron transport chain (ETC) Complex I, II, and IV] were compared between PBBI and sham groups. Furthermore, mitochondrial coenzyme contents, including NAD(t) and FAD(t), were quantitatively measured in both groups. Collectively, PBBI led to an overall significant decline in the ATP synthesis rates (43-50%; * p < 0.05 vs. sham) when measured using each of the four sets of substrates. The PDHC and GDH activities were significantly reduced in the PBBI group (42-53%; * p < 0.05 vs. sham), whereas no significant differences were noted in α-KGDHC activity between groups. Both Complex I and Complex IV activities were significantly reduced following PBBI (47-81%; * p < 0.05 vs. sham), whereas, Complex II activity was comparable between groups. The NAD(t) and FAD(t) contents were significantly decreased in the PBBI group (27-35%; * p < 0.05 vs. sham). The decreased ATP synthesis rates may be due to the significant reductions in brain mitochondrial dehydrogenase activities and coenzyme contents observed acutely following PBBI. These results provide a basis for the use of "alternative biofuels" for achieving higher ATP production following severe penetrating brain trauma.
Collapse
Affiliation(s)
- Jignesh D Pandya
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Lai Yee Leung
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Xiaofang Yang
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - William J Flerlage
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Ying Deng-Bryant
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah A Shear
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
14
|
Nicotinamide Improves Functional Recovery via Regulation of the RAGE/JNK/NF-κB Signaling Pathway after Brain Injury. J Clin Med 2019; 8:jcm8020271. [PMID: 30813383 PMCID: PMC6406790 DOI: 10.3390/jcm8020271] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022] Open
Abstract
Brain injuries are a serious global health issue and are the leading cause of neurodegeneration. To date, there is no proper cure and treatment for brain-injury-induced neuropathological conditions because of a lack of sufficient knowledge and the failure to develop a drug due to the multi-pathological conditions in the brain. Herein, we explored the neurotherapeutic effects of Nicotinamide (NAM), against brain injury-induced neurodegeneration and behavioral problems. Treating injured mouse brains with NAM, for 7 days, significantly ameliorated several pathological events. Interestingly, NAM treatment significantly inhibited the injury-induced activation of receptor for advanced glycation end-products (RAGE), c-Jun N-terminal kinases (JNK), and neuroinflammatory mediators, such as NF-κB, TNF-α, IL-1β, and NOS2 in the brain, and it also regulated the levels of apoptotic markers, including Bax, caspase-3, and Bcl-2. Furthermore, treatment using NAM in TBI mice, significantly reversed synaptic protein loss and improved memory impairments and behavioral outcomes. Our findings suggested that NAM treatment reduced injury-induced secondary neurodegenerative pathology by modulating RAGE/JNK/NF-κB signaling in mice. Therefore, we recommend that NAM would be a safe and efficient therapeutic agent against brain-injury-induced neurodegeneration.
Collapse
|
15
|
Smith AC, Holden RC, Rasmussen SM, Hoane MR, Hylin MJ. Effects of nicotinamide on spatial memory and inflammation after juvenile traumatic brain injury. Behav Brain Res 2019; 364:123-132. [PMID: 30771366 DOI: 10.1016/j.bbr.2019.02.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 01/23/2023]
Abstract
Age is a consistent predictor of outcome following traumatic brain injury (TBI). Although children and adolescents have the highest rate of hospitalizations and long-term disabilities, few preclinical studies have attempted to model and treat TBI in this population. Studies using nicotinamide (NAM), a soluble B-group vitamin, in older animals (3-6 months) have shown improved functional recovery in experimental models of TBI. The purpose of this study was two-fold: to examine the preclinical efficacy of NAM at different doses on behavioral outcomes in juvenile rats and examine the microglial response over time. Groups of juvenile rats (PND 28-60) were assigned to sham, NAM (125 mg/kg, 500 mg/kg, or 1000 mg/kg) or saline (1 mL/kg) and received unilateral cortical contusion injuries (CCI) and received injections at 15 min, 24 h, and 72 h after injury. Animals treated with NAM demonstrated no significant behavioral improvements over saline treatments. NAM treatments did however show slowed cortical loss and reduced microglia compared to saline treated animals. In summary, the preclinical efficacy of NAM as a treatment following CCI in juvenile animals differs from that previously documented in older rat models. While NAM treatments did reduce microglial activity and slowed progression of cortical loss, it did not reduce the total cortical volume lost nor did it improve behavioral outcomes. The findings of this study emphasize the need to examine potential treatments for TBI utilizing juvenile populations and may explain why so many treatments have failed in clinical trials.
Collapse
Affiliation(s)
- Aidan C Smith
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Ryan C Holden
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Sherry M Rasmussen
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Michael R Hoane
- Restorative Neuroscience Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Michael J Hylin
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States.
| |
Collapse
|
16
|
Tóth L, Szöllősi D, Kis-Petik K, Adorján I, Erdélyi F, Kálmán M. The First Postlesion Minutes: An In Vivo Study of Extravasation and Perivascular Astrocytes Following Cerebral Lesions in Various Experimental Mouse Models. J Histochem Cytochem 2018; 67:29-39. [PMID: 30047826 DOI: 10.1369/0022155418788390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The immediate alterations following lesions cannot be investigated by using fixed tissues. Here, we employed two-photon microscopy to study the alterations to the permeability of blood-brain barrier and to glio-vascular connections in vivo during the first minutes following cortical lesions in mice. Four models were used: (1) cryogenic lesion, (2) photodisruption using laser pulses, (3) photothrombosis, and (4) bilateral carotid ligation. Sulforhodamine101 was used for supravital labeling of astrocytes and dextran-bound fluorescein isothiocyanate for the assessment of extravasation. Transgenic mice, in which the endothelium and astrocytes expressed a yellow fluorescent protein, were also used. Astrocytic labeling in vivo was verified with postmortem immunostaining against glial fibrillary acidic protein (GFAP). Summary of results: (1) the glio-vascular connections were stable in the intact brain with no sign of spontaneous dynamic attachment/detachment of glial end-feet; (2) only direct vascular damage (photodisruption or cryogenic) resulted in prompt extravasation; (3) even direct damage failed to provoke a prompt astroglial response. In conclusion, the results indicate that a detachment of the astrocytic end-feet does not precede the breakdown of blood-brain barrier following lesions. Whereas vasogenic edema develops immediately after the lesions, this is not the case with cytotoxic edemas. Time-lapse recordings and three-dimensional reconstructions are presented as supplemental materials.
Collapse
Affiliation(s)
- László Tóth
- Department of Anatomy, Histology and Embryology (LT, DS, IA, MK).,Department of Biophysics and Radiation Biology, MTA-SE Molecular Biology Research Group (DS, KK-P).,Semmelweis University, Budapest, Hungary, and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (FE)
| | - Dávid Szöllősi
- Department of Anatomy, Histology and Embryology (LT, DS, IA, MK).,Department of Biophysics and Radiation Biology, MTA-SE Molecular Biology Research Group (DS, KK-P).,Semmelweis University, Budapest, Hungary, and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (FE)
| | - Katalin Kis-Petik
- Department of Anatomy, Histology and Embryology (LT, DS, IA, MK).,Department of Biophysics and Radiation Biology, MTA-SE Molecular Biology Research Group (DS, KK-P).,Semmelweis University, Budapest, Hungary, and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (FE)
| | - István Adorján
- Department of Anatomy, Histology and Embryology (LT, DS, IA, MK).,Department of Biophysics and Radiation Biology, MTA-SE Molecular Biology Research Group (DS, KK-P).,Semmelweis University, Budapest, Hungary, and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (FE)
| | - Ferenc Erdélyi
- Department of Anatomy, Histology and Embryology (LT, DS, IA, MK).,Department of Biophysics and Radiation Biology, MTA-SE Molecular Biology Research Group (DS, KK-P).,Semmelweis University, Budapest, Hungary, and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (FE)
| | - Mihály Kálmán
- Department of Anatomy, Histology and Embryology (LT, DS, IA, MK).,Department of Biophysics and Radiation Biology, MTA-SE Molecular Biology Research Group (DS, KK-P).,Semmelweis University, Budapest, Hungary, and Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary (FE)
| |
Collapse
|
17
|
Effects of Protocatechuic Acid (PCA) on Global Cerebral Ischemia-Induced Hippocampal Neuronal Death. Int J Mol Sci 2018; 19:ijms19051420. [PMID: 29747437 PMCID: PMC5983751 DOI: 10.3390/ijms19051420] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
Global cerebral ischemia (GCI) is one of the main causes of hippocampal neuronal death. Ischemic damage can be rescued by early blood reperfusion. However, under some circumstances reperfusion itself can trigger a cell death process that is initiated by the reintroduction of blood, followed by the production of superoxide, a blood⁻brain barrier (BBB) disruption and microglial activation. Protocatechuic acid (PCA) is a major metabolite of the antioxidant polyphenols, which have been discovered in green tea. PCA has been shown to have antioxidant effects on healthy cells and anti-proliferative effects on tumor cells. To test whether PCA can prevent ischemia-induced hippocampal neuronal death, rats were injected with PCA (30 mg/kg/day) per oral (p.o) for one week after global ischemia. To evaluate degenerating neurons, oxidative stress, microglial activation and BBB disruption, we performed Fluoro-Jade B (FJB), 4-hydroxynonenal (4HNE), CD11b, GFAP and IgG staining. In the present study, we found that PCA significantly decreased degenerating neuronal cell death, oxidative stress, microglial activation, astrocyte activation and BBB disruption compared with the vehicle-treated group after ischemia. In addition, an ischemia-induced reduction in glutathione (GSH) concentration in hippocampal neurons was recovered by PCA administration. Therefore, the administration of PCA may be further investigated as a promising tool for decreasing hippocampal neuronal death after global cerebral ischemia.
Collapse
|
18
|
Progress in the treatment of Friedreich ataxia. Neurol Neurochir Pol 2018; 52:129-139. [PMID: 29499876 DOI: 10.1016/j.pjnns.2018.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/12/2018] [Indexed: 11/19/2022]
Abstract
Friedreich ataxia (FRDA) is a progressive neurological disorder affecting approximately 1 in 29,000 individuals of European descent. At present, there is no approved pharmacological treatment for this condition however research into treatment of FRDA has advanced considerably over the last two decades since the genetic cause was identified. Current proposed treatment strategies include decreasing oxidative stress, increasing cellular frataxin, improving mitochondrial function as well as modulating frataxin controlled metabolic pathways. Genetic and cell based therapies also hold great promise. Finally, physical therapies are being explored as a means of maximising function in those affected by FRDA.
Collapse
|
19
|
Kálmán M, Tóth L, Szöllosi D, Oszwald E, Mahalek J, Sadeghian S. Correlation Between Extravasation and Alterations of Cerebrovascular Laminin and β-Dystroglycan Immunoreactivity Following Cryogenic Lesions in Rats. J Neuropathol Exp Neurol 2017; 76:929-941. [PMID: 29044412 DOI: 10.1093/jnen/nlx081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The blood-brain barrier becomes "leaky" following lesions. Former studies revealed that following lesions the immunoreactivity of cerebrovascular laminin becomes detectable whereas that of β-dystroglycan disappears. These alterations may be indicators of glio-vascular decoupling that may result in the impairment of the blood-brain-barrier. This study investigates correlation between the post-lesion extravasation and the above-mentioned immunohistochemical alterations. Following cryogenic lesions, the survival periods lasted 5, 10, 30 minutes, 1 or 12 hours, or 1 day. Some brains were fixed immediately post-lesion. Immunofluorescent reactions were performed in floating sections. The extravasation was detected with immunostaining for plasma fibronectin and rat immunoglobulins. When the survival period was 30 minutes or longer, the area of extravasation corresponded to the area of altered laminin and β-dystroglycan immunoreactivities. Following immediate fixation some laminin immunoreactivity was already detected. The extravasation seemed to precede this early appearance of laminin immunoreactivity. The β-dystroglycan immunoreactivity disappeared later. When the extravasation spread into the corpus callosum, vascular laminin immunoreactivity appeared but the β-dystroglycan immunoreactivity persisted. It seems that extravasation separates the glial and vascular basal laminae, which results in the appearance of laminin immunoreactivity. The disappearance of β-dystroglycan immunoreactivity is neither a condition nor an inevitable consequence of the 2 other phenomena.
Collapse
Affiliation(s)
- Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - László Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Dávid Szöllosi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Oszwald
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Judit Mahalek
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Sam Sadeghian
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Drake DF, Hudak AM, Robbins W. Integrative Medicine in Traumatic Brain Injury. Phys Med Rehabil Clin N Am 2017; 28:363-378. [DOI: 10.1016/j.pmr.2016.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Shear DA, Dixon CE, Bramlett HM, Mondello S, Dietrich WD, Deng-Bryant Y, Schmid KE, Wang KKW, Hayes RL, Povlishock JT, Kochanek PM, Tortella FC. Nicotinamide Treatment in Traumatic Brain Injury: Operation Brain Trauma Therapy. J Neurotrauma 2016; 33:523-37. [PMID: 26670792 DOI: 10.1089/neu.2015.4115] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nicotinamide (vitamin B3) was the first drug selected for cross-model testing by the Operation Brain Trauma Therapy (OBTT) consortium based on a compelling record of positive results in pre-clinical models of traumatic brain injury (TBI). Adult male Sprague-Dawley rats were exposed to either moderate fluid percussion injury (FPI), controlled cortical impact injury (CCI), or penetrating ballistic-like brain injury (PBBI). Nicotinamide (50 or 500 mg/kg) was delivered intravenously at 15 min and 24 h after injury with subsequent behavioral, biomarker, and histopathological outcome assessments. There was an intermediate effect on balance beam performance with the high (500 mg/kg) dose in the CCI model, but no significant therapeutic benefit was detected on any other motor task across the OBTT TBI models. There was an intermediate benefit on working memory with the high dose in the FPI model. A negative effect of the low (50 mg/kg) dose, however, was observed on cognitive outcome in the CCI model, and no cognitive improvement was observed in the PBBI model. Lesion volume analysis showed no treatment effects after either FPI or PBBI, but the high dose of nicotinamide resulted in significant tissue sparing in the CCI model. Biomarker assessments included measurements of glial fibrillary acidic protein (GFAP) and ubiquitin carboxyl-terminal hydrolase-1 (UCH-L1) in blood at 4 or 24 h after injury. Negative effects (both doses) were detected on biomarker levels of GFAP after FPI and on biomarker levels of UCH-L1 after PBBI. The high dose of nicotinamide, however, reduced GFAP levels after both PBBI and CCI. Overall, our results showed a surprising lack of benefit from the low dose nicotinamide. In contrast, and partly in keeping with the literature, some benefit was achieved with the high dose. The marginal benefits achieved with nicotinamide, however, which appeared sporadically across the TBI models, has reduced enthusiasm for further investigation by the OBTT Consortium.
Collapse
Affiliation(s)
- Deborah A Shear
- 1 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - C Edward Dixon
- 2 Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Helen M Bramlett
- 3 Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami , Miami, Florida.,4 Bruce W. Carter Department of Veterans Affairs Medical Center , Miami, Florida
| | - Stefania Mondello
- 5 Department of Neurosciences, University of Messina , Messina, Italy
| | - W Dalton Dietrich
- 3 Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami , Miami, Florida
| | - Ying Deng-Bryant
- 1 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Kara E Schmid
- 1 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Kevin K W Wang
- 6 Center of Neuroproteomics and Biomarkers Research, Department of Psychiatry and Neuroscience, University of Florida , Gainesville, Florida
| | - Ronald L Hayes
- 7 Center for Innovative Research, Center for Neuroproteomics and Biomarkers Research , Banyan Biomarkers, Inc., Alachua, Florida
| | - John T Povlishock
- 8 Department of Anatomy and Neurobiology, Virginia Commonwealth University , Richmond, Virginia
| | - Patrick M Kochanek
- 9 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Frank C Tortella
- 1 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| |
Collapse
|
22
|
Vonder Haar C, Peterson TC, Martens KM, Hoane MR. Vitamins and nutrients as primary treatments in experimental brain injury: Clinical implications for nutraceutical therapies. Brain Res 2016; 1640:114-129. [PMID: 26723564 PMCID: PMC4870112 DOI: 10.1016/j.brainres.2015.12.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
With the numerous failures of pharmaceuticals to treat traumatic brain injury in humans, more researchers have become interested in combination therapies. This is largely due to the multimodal nature of damage from injury, which causes excitotoxicity, oxidative stress, edema, neuroinflammation and cell death. Polydrug treatments have the potential to target multiple aspects of the secondary injury cascade, while many previous therapies focused on one particular aspect. Of specific note are vitamins, minerals and nutrients that can be utilized to supplement other therapies. Many of these have low toxicity, are already FDA approved and have minimal interactions with other drugs, making them attractive targets for therapeutics. Over the past 20 years, interest in supplementation and supraphysiologic dosing of nutrients for brain injury has increased and indeed many vitamins and nutrients now have a considerable body of the literature backing their use. Here, we review several of the prominent therapies in the category of nutraceutical treatment for brain injury in experimental models, including vitamins (B2, B3, B6, B9, C, D, E), herbs and traditional medicines (ginseng, Gingko biloba), flavonoids, and other nutrients (magnesium, zinc, carnitine, omega-3 fatty acids). While there is still much work to be done, several of these have strong potential for clinical therapies, particularly with regard to polydrug regimens. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
|
23
|
Zhang L, Ding K, Wang H, Wu Y, Xu J. Traumatic Brain Injury-Induced Neuronal Apoptosis is Reduced Through Modulation of PI3K and Autophagy Pathways in Mouse by FTY720. Cell Mol Neurobiol 2016; 36:131-42. [PMID: 26099903 PMCID: PMC11482378 DOI: 10.1007/s10571-015-0227-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/12/2015] [Indexed: 01/13/2023]
Abstract
FTY720 is a synthetic compound produced by modification of metabolite from Isaria sinclairii. It is a novel type of immunosuppressive agent inhibiting lymphocyte egress from secondary lymphoid tissues, thereby causing peripheral lymphopenia. Growing evidences have suggested that apoptosis and autophagy were involved in the secondary brain injury after traumatic brain injury (TBI) although FTY720 exerted neuroprotective effects in a variety of neurological diseases except TBI. The present study was aimed to investigate the role of FTY720 in a mouse model of TBI. In experiment 1, ICR mice were divided into four groups: sham group, TBI group, TBI + vehicle group, and TBI + FTY720 group. And the injured cerebral cortex (including both contused and penumbra) was used for analysis. We found that FTY720 administration after TBI improved neurobehavioral function, alleviated brain edema, accompanied by modulation of apoptotic indicators such as Bcl-2, Bcl-xL, Bax, and cytochrome c. In experiment 2, ICR mice were also divided into four groups: sham group, TBI + vehicle group, TBI + FTY720 group, and TBI + FTY720 + inhibitors group. And the injured cerebral cortex (including both contused and penumbra) was used for analysis. We found that FTY720 increased the expression of phospho-protein kinase B (AKT) and some autophagy markers such as LC3 and Beclin 1. In addition, the apoptosis inhibition effect of FTY720 was partly abrogated by the phosphatidylinositide 3-kinases (PI3K)/AKT pathway inhibitor LY294002 and autophagy inhibitor 3-methyladenine. Collectively, our data provide the first evidence that FTY720 exerted neuroprotective effects after TBI, at least in part, through the activation of PI3K/AKT pathway and autophagy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Yong Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Jianguo Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| |
Collapse
|
24
|
Peterson TC, Hoane MR, McConomy KS, Farin FM, Bammler TK, MacDonald JW, Kantor ED, Anderson GD. A Combination Therapy of Nicotinamide and Progesterone Improves Functional Recovery following Traumatic Brain Injury. J Neurotrauma 2015; 32:765-79. [PMID: 25313690 DOI: 10.1089/neu.2014.3530] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuroprotection, recovery of function, and gene expression were evaluated in an animal model of traumatic brain injury (TBI) after a combination treatment of nicotinamide (NAM) and progesterone (Prog). Animals received a cortical contusion injury over the sensorimotor cortex, and were treated with either Vehicle, NAM, Prog, or a NAM/Prog combination for 72 h and compared with a craniotomy only (Sham) group. Animals were assessed in a battery of behavioral, sensory, and both fine and gross motor tasks, and given histological assessments at 24 h post-injury to determine lesion cavity size, degenerating neurons, and reactive astrocytes. Microarray-based transcriptional profiling was used to determine treatment-specific changes on gene expression. Our results confirm the beneficial effects of treatment with either NAM or Prog, demonstrating significant improvements in recovery of function and a reduction in lesion cavitation, degenerating neurons, and reactive astrocytes 24 h post-injury. The combination treatment of NAM and Prog led to a significant improvement in both neuroprotection at 24 h post-injury and recovery of function in sensorimotor related tasks when compared with individual treatments. The NAM/Prog-treated group was the only treatment group to show a significant reduction of cortical loss 24 h post-injury. The combination appears to affect inflammatory and immune processes, reducing expression of a significant number of genes in both pathways. Further preclinical trials using NAM and Prog as a combination treatment should be conducted to identify the window of opportunity, determine the optimal duration of treatment, and evaluate the combination in other pre-clinical models of TBI.
Collapse
Affiliation(s)
- Todd C Peterson
- 1Department of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Michael R Hoane
- 1Department of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Keith S McConomy
- 1Department of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Fred M Farin
- 2Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Theo K Bammler
- 2Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - James W MacDonald
- 2Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Eric D Kantor
- 3Department of Pharmacy, University of Washington, Seattle, Washington
| | - Gail D Anderson
- 3Department of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
25
|
Nicotinamide induces apoptosis of F9 mouse teratocarcinoma stem cells by downregulation of SATB1 expression. Tumour Biol 2015; 36:4339-48. [PMID: 25596087 DOI: 10.1007/s13277-015-3073-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/08/2015] [Indexed: 10/24/2022] Open
Abstract
The aim of this study was to decide whether nicotinamide (NA) could induce apoptosis of F9 mouse teratocarcinoma stem cells (MF9) by downregulation of special AT-rich sequence binding protein 1 (SATB1) expression. We used different concentrations of NA (0, 1.5, 2, and 2.5 mmol/L) to treat MF9 cells and analyze SATB1 expression by RT-qPCR and Western blotting; in addition, the cell proliferation was detected in a microplate reader with Cell Counting Kit-8 (CCK-8), and the cell cycle and apoptosis were analyzed using flow cytometry. We found that the expression of SATB1 was decreased significantly in NA-treated groups than in the control group, and its expression level was inversely related to the NA concentration. In addition, CCK-8 analysis showed that NA significantly inhibited the proliferation of MF9 cells, and flow cytometry showed that NA blocked MF9 cells to G1 phase and significantly promoted apoptosis in any treated groups. To confirm the results, we constructed small interference RNA (siRNA) targeting at mouse SATB1 and transferred into MF9 cells. The results indicated that the expression of SATB1 in both mRNA and protein levels was significantly decreased after cells transferred with siRNA sequence for 48 h, the proliferation of MF9 cells was significantly inhibited, and most of MF9 cells were blocked at G1 phase, and the apoptosis rate was increased obviously. The results showed that NA could inhibit the proliferation and induce apoptosis of MF9 cells. These findings might be used as an efficient candidate for teratocarcinoma therapy.
Collapse
|
26
|
Libri V, Yandim C, Athanasopoulos S, Loyse N, Natisvili T, Law PP, Chan PK, Mohammad T, Mauri M, Tam KT, Leiper J, Piper S, Ramesh A, Parkinson MH, Huson L, Giunti P, Festenstein R. Epigenetic and neurological effects and safety of high-dose nicotinamide in patients with Friedreich's ataxia: an exploratory, open-label, dose-escalation study. Lancet 2014; 384:504-13. [PMID: 24794816 DOI: 10.1016/s0140-6736(14)60382-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Friedreich's ataxia is a progressive degenerative disorder caused by deficiency of the frataxin protein. Expanded GAA repeats within intron 1 of the frataxin (FXN) gene lead to its heterochromatinisation and transcriptional silencing. Preclinical studies have shown that the histone deacetylase inhibitor nicotinamide (vitamin B3) can remodel the pathological heterochromatin and upregulate expression of FXN. We aimed to assess the epigenetic and neurological effects and safety of high-dose nicotinamide in patients with Friedreich's ataxia. METHODS In this exploratory, open-label, dose-escalation study in the UK, male and female patients (aged 18 years or older) with Friedreich's ataxia were given single doses (phase 1) and repeated daily doses of 2-8 g oral nicotinamide for 5 days (phase 2) and 8 weeks (phase 3). Doses were gradually escalated during phases 1 and 2, with individual maximum tolerated doses used in phase 3. The primary outcome was the upregulation of frataxin expression. We also assessed the safety and tolerability of nicotinamide, used chromatin immunoprecipitation to investigate changes in chromatin structure at the FXN gene locus, and assessed the effect of nicotinamide treatment on clinical scales for ataxia. This study is registered with ClinicalTrials.gov, number NCT01589809. FINDINGS Nicotinamide was generally well tolerated; the main adverse event was nausea, which in most cases was mild, dose-related, and resolved spontaneously or after dose reduction, use of antinausea drugs, or both. Phase 1 showed a dose-response relation for proportional change in frataxin protein concentration from baseline to 8 h post-dose, which increased with increasing dose (p=0·0004). Bayesian analysis predicted that 3·8 g would result in a 1·5-times increase and 7·5 g in a doubling of frataxin protein concentration. Phases 2 and 3 showed that daily dosing at 3·5-6 g resulted in a sustained and significant (p<0·0001) upregulation of frataxin expression, which was accompanied by a reduction in heterochromatin modifications at the FXN locus. Clinical measures showed no significant changes. INTERPRETATION Nicotinamide was associated with a sustained improvement in frataxin concentrations towards those seen in asymptomatic carriers during 8 weeks of daily dosing. Further investigation of the long-term clinical benefits of nicotinamide and its ability to ameliorate frataxin deficiency in Friedreich's ataxia is warranted. FUNDING Ataxia UK, Ataxia Ireland, Association Suisse de l'Ataxie de Friedreich, Associazione Italiana per le Sindromi Atassiche, UK National Institute for Health Research, European Friedreich's Ataxia Consortium for Translational Studies, and Imperial Biomedical Research Centre.
Collapse
Affiliation(s)
- Vincenzo Libri
- Leonard Wolfson Experimental Neurology Centre, University College London, London, UK; National Institute for Health Research Wellcome Trust Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Cihangir Yandim
- Gene Control Mechanisms and Disease Group, Department of Medicine and MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Stavros Athanasopoulos
- National Institute for Health Research Wellcome Trust Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Naomi Loyse
- National Institute for Health Research Wellcome Trust Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Theona Natisvili
- Gene Control Mechanisms and Disease Group, Department of Medicine and MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Pui Pik Law
- Gene Control Mechanisms and Disease Group, Department of Medicine and MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Ping Kei Chan
- Gene Control Mechanisms and Disease Group, Department of Medicine and MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Tariq Mohammad
- National Institute for Health Research Wellcome Trust Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Marta Mauri
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Kin Tung Tam
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - James Leiper
- Nitric Oxide Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Sophie Piper
- Nitric Oxide Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK
| | - Aravind Ramesh
- Intensive Care Department, Christchurch Hospital, Christchurch, New Zealand
| | - Michael H Parkinson
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Les Huson
- National Institute for Health Research Wellcome Trust Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Paola Giunti
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Richard Festenstein
- Gene Control Mechanisms and Disease Group, Department of Medicine and MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
27
|
Torres-Ramírez N, Baiza-Gutman LA, García-Macedo R, Ortega-Camarillo C, Contreras-Ramos A, Medina-Navarro R, Cruz M, Ibáñez-Hernández MÁ, Díaz-Flores M. Nicotinamide, a glucose-6-phosphate dehydrogenase non-competitive mixed inhibitor, modifies redox balance and lipid accumulation in 3T3-L1 cells. Life Sci 2013; 93:975-85. [PMID: 24184296 DOI: 10.1016/j.lfs.2013.10.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/30/2013] [Accepted: 10/19/2013] [Indexed: 01/21/2023]
Abstract
AIMS Excessive energy uptake of dietary carbohydrates results in their storage as fat and requires glucose-6-phosphate dehydrogenase (G6PD)-mediated NADPH production. We sought to assess whether the nicotinamide-induced reduction of G6PD activity might modulate redox balance and lipid accumulation in 3T3-L1 cells. MAIN METHODS 3T3-L1 preadipocytes (days 4 and 6 of differentiation) and adipocytes were cultured in the presence of 5 or 25 mM glucose. The cells cultured in 25 mM glucose were supplemented with nicotinamide (5-15 mM). Next, we evaluated the following parameters: cell viability, apoptosis, lipid accumulation, lipolysis, reducing power, reactive oxygen species (ROS), NAD(P)H and NAD(P)(+), isocitrate dehydrogenase (IDP), malic enzyme and G6PD, as well as the protein and mRNA levels of G6PD. We also analysed the kinetics of the nicotinamide-induced inhibition of G6PD. KEY FINDINGS G6PD mRNA levels increased at day 4 of adipocyte differentiation, whereas G6PD activity progressively increased at days 4 and 6 of differentiation and was reduced in adipocytes. Concomitantly, ROS, reducing power and lipid accumulation increased gradually as the preadipocytes matured into adipocytes. High glucose increased the activity of G6PD, which coincided with an increase in ROS, reducing power and lipid accumulation. All of these changes are prevented by nicotinamide, with the exception of lipid accumulation in adipocytes. Nicotinamide increased IDP activity without affecting NADPH levels. Lastly, nicotinamide inhibited G6PD in a non-competitive mixed way. SIGNIFICANCE Nicotinamide modulates G6PD via a non-competitive mixed inhibition and decreases high glucose-dependent oxidative stress and lipid accumulation. Nicotinamide maintains NADPH levels by increasing the activity of IDP.
Collapse
Affiliation(s)
- Nayeli Torres-Ramírez
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, CMN Siglo XXI, IMSS, D.F., Mexico; Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, D.F., Mexico; Posgrado en Biomedicina y Biotecnología Molecular, ENCB-Instituto Politécnico Nacional, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yu CG, Singh R, Crowdus C, Raza K, Kincer J, Geddes JW. Fenbendazole improves pathological and functional recovery following traumatic spinal cord injury. Neuroscience 2013; 256:163-9. [PMID: 24183965 DOI: 10.1016/j.neuroscience.2013.10.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/17/2013] [Accepted: 10/19/2013] [Indexed: 01/16/2023]
Abstract
During a study of spinal cord injury (SCI), mice in our colony were treated with the anthelmintic fenbendazole to treat pinworms detected in other mice not involved in the study. As this was not part of the original experimental design, we subsequently compared pathological and functional outcomes of SCI in female C57BL/6 mice who received fenbendazole (150 ppm, 8 mg/kg body weight/day) for 4 weeks prior to moderate contusive SCI (50 kdyn force) as compared to mice on the same diet without added fenbendazole. The fenbendazole-treated mice exhibited improved locomotor function, determined using the Basso mouse scale, as well as improved tissue sparing following contusive SCI. Fenbendazole may exert protective effects through multiple possible mechanisms, one of which is inhibition of the proliferation of B lymphocytes, thereby reducing antibody responses. Autoantibodies produced following SCI contribute to the axon damage and locomotor deficits. Fenbendazole pretreatment reduced the injury-induced CD45R-positive B cell signal intensity and IgG immunoreactivity at the lesion epicenter 6 weeks after contusive SCI in mice, consistent with a possible effect on the immune response to the injury. Fenbendazole and related benzimadole antihelmintics are FDA approved, exhibit minimal toxicity, and represent a novel group of potential therapeutics targeting secondary mechanisms following SCI.
Collapse
Affiliation(s)
- C G Yu
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - R Singh
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - C Crowdus
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - K Raza
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - J Kincer
- Division of Laboratory Animal Resources, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - J W Geddes
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| |
Collapse
|
29
|
Koh PO. Nicotinamide attenuates the injury-induced decrease of hippocalcin in ischemic brain injury. Neurosci Lett 2013; 545:6-10. [DOI: 10.1016/j.neulet.2013.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 04/01/2013] [Accepted: 04/02/2013] [Indexed: 11/25/2022]
|
30
|
Anderson GD, Peterson TC, Farin FM, Bammler TK, Beyer RP, Kantor ED, Hoane MR. The effect of nicotinamide on gene expression in a traumatic brain injury model. Front Neurosci 2013; 7:21. [PMID: 23550224 PMCID: PMC3581799 DOI: 10.3389/fnins.2013.00021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/06/2013] [Indexed: 12/30/2022] Open
Abstract
Microarray-based transcriptional profiling was used to determine the effect of nicotinamide on gene expression in an experimental traumatic brain injury (TBI) model. Ingenuity Pathway Analysis (IPA) was used to evaluate the effect on relevant functional categories and canonical pathways. At 24 h, 72 h, and 7 days, respectively, 70, 58, and 76%, of the differentially expressed genes were up-regulated in the vehicle treated compared to the sham animals. At 24 h post-TBI, there were 150 differentially expressed genes in the nicotinamide treated animals compared to vehicle; the majority (82%) down-regulated. IPA analysis identified a significant effect of nicotinamide on the functional categories of cellular movement, cell-to-cell-signaling, antigen presentation and cellular compromise, function, and maintenance and cell death. The canonical pathways identified were signaling pathways primarily involved with the inflammatory process. At 72 h post-cortical contusion injury, there were 119 differentially expressed genes in the nicotinamide treated animals compared to vehicle; the majority (90%) was up-regulated. IPA analysis identified a significant effect of nicotinamide on cell signaling pathways involving neurotransmitters, neuropeptides, growth factors, and ion channels with little to no effect on inflammatory pathways. At 7 days post-TBI, there were only five differentially expressed genes with nicotinamide treatment compared to vehicle. Overall, the effect of nicotinamide on counteracting the effect of TBI resulted in significantly decreased number of genes differentially expressed by TBI. In conclusion, the mechanism of the effect of nicotinamide on secondary injury pathways involves effects on inflammatory response, signaling pathways, and cell death.
Collapse
Affiliation(s)
- G D Anderson
- Department of Pharmacy, University of Washington Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Cantó C, Sauve AA, Bai P. Crosstalk between poly(ADP-ribose) polymerase and sirtuin enzymes. Mol Aspects Med 2013; 34:1168-201. [PMID: 23357756 DOI: 10.1016/j.mam.2013.01.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/07/2013] [Accepted: 01/17/2013] [Indexed: 01/08/2023]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are NAD(+) dependent enzymes that were identified as DNA repair proteins, however, today it seems clear that PARPs are responsible for a plethora of biological functions. Sirtuins (SIRTs) are NAD(+)-dependent deacetylase enzymes involved in the same biological processes as PARPs raising the question whether PARP and SIRT enzymes may interact with each other in physiological and pathophysiological conditions. Hereby we review the current understanding of the SIRT-PARP interplay in regard to the biochemical nature of the interaction (competition for the common NAD(+) substrate, mutual posttranslational modifications and direct transcriptional effects) and the physiological or pathophysiological consequences of the interactions (metabolic events, oxidative stress response, genomic stability and aging). Finally, we give an overview of the possibilities of pharmacological intervention to modulate PARP and SIRT enzymes either directly, or through modulating NAD(+) homeostasis.
Collapse
Affiliation(s)
- Carles Cantó
- Nestlé Institute of Health Sciences, Lausanne CH-1015, Switzerland
| | | | | |
Collapse
|
32
|
KOH PO. Nicotinamide Restores the Reduction of Parvalbumin in Cerebral Ischemic Injury. J Vet Med Sci 2013; 75:225-9. [DOI: 10.1292/jvms.12-0216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Phil-Ok KOH
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 900 Gajwa-dong, Jinju 660–701, South Korea
| |
Collapse
|
33
|
Huang XJ, Mao Q, Lin Y, Feng JF, Jiang JY. Expression of voltage-gated sodium channel Nav1.3 is associated with severity of traumatic brain injury in adult rats. J Neurotrauma 2012; 30:39-46. [PMID: 22928478 DOI: 10.1089/neu.2012.2508] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
During the secondary injury period after traumatic brain injury (TBI), depolarization of neurons mediated by voltage-gated sodium channels (VGSCs) leads to cellular abnormalities and neurological dysfunction. Alterations in expression of different α subunits of VGSCs can affect early brain pathology following TBI. This study detected the expression of Nav1.3 mRNA and protein in the rat cortex post-TBI. Adult male Sprague-Dawley rats were randomly assigned to sham-TBI, mild-TBI (mTBI), or severe-TBI (sTBI) groups. TBI was induced using a fluid percussion device at magnitudes of 1.5-1.6 atm (mTBI) and 2.9-3.0 atm (sTBI). Nav1.3 mRNA and protein levels in the ipsilateral-injured cortex were examined at 2 h, 12 h, 24 h, and 72 h post-TBI by real-time reverse transcriptase quantitative polymerase chain reaction and Western blot. Brains were collected at 24 h, 72 h, and 7 days post-TBI for TUNEL staining and cell count analysis. Immunofluorescence was performed to localize expression of Nav1.3 protein in the ipsilateral-injured cortex. Expression of Nav1.3 mRNA and protein were significantly upregulated in mTBI and sTBI groups when compared with the sham-TBI group at 2 h and 12 h post-TBI. Nav1.3 mRNA and protein levels in the sTBI group were much higher than in the mTBI group at 12 h post-TBI. TUNEL-positive cell numbers were significantly higher in the sTBI group than in the mTBI at 24 h, 72 h, and 7 days post-TBI. Expression of Nav1.3 was observed predominantly in neurons of the cortex. These findings indicated significant upregulation in the expression of Nav1.3 mRNA and protein in the rat ipsilateral-injured cortex at the very early stage post-TBI, and were also correlated with TBI severity.
Collapse
Affiliation(s)
- Xian-jian Huang
- Department of Neurosurgery, Shanghai Renji Hospital, Shanghai Jiaotong University , School of Medicine, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
34
|
Peterson TC, Anderson GD, Kantor ED, Hoane MR. A comparison of the effects of nicotinamide and progesterone on functional recovery of cognitive behavior following cortical contusion injury in the rat. J Neurotrauma 2012; 29:2823-30. [PMID: 23016598 PMCID: PMC3521133 DOI: 10.1089/neu.2012.2471] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The primary goal of this study was to compare clinically relevant doses of progesterone and nicotinamide within the same injury model. Progesterone has been shown to reduce edema and inflammation and improve functional outcomes following brain injury. Nicotinamide has also been shown to be an effective neuroprotective agent in a variety of neurological injury models. In the current study, nicotinamide was administered beginning 4 h post-cortical contusion injury (CCI) with a loading dose (75 mg/kg, i.p.) combined with continuous infusion (12 mg/h/kg, s.c.) for 72 h post-injury. Progesterone was administered beginning 4 h post-CCI at a dose of 10 or 20 mg/kg, i.p. every 12 h for 72 h. This resulted in the following groups: Injured-nicotinamide treated, Injured-progesterone-10 treated, Injured-progesterone-20 treated, Injured-vehicle treated, and Sham. Functional recovery was assessed with two spatial memory tasks in the Morris water maze (MWM) the acquisition of a reference memory task and a reversal learning task. Neuropathological assessments were conducted in the cortex and hippocampus. It was found that both progesterone (10 mg/kg) and nicotinamide improved reference memory acquisition and reversal learning in the MWM compared with vehicle treatment. The lower dose of progesterone and nicotinamide also reduced tissue loss in the injured cortex and ipsilateral hippocampus compared with vehicle. The beneficial effects of progesterone appear to be dose dependent with the lower 10 mg/kg dose producing significant effects that were not observed at the higher dose. Direct comparison between nicotinamide and low dose progesterone appears to suggest that both are equally effective. The general findings of this study suggest that both nicotinamide and progesterone produce significant improvements in recovery of function following CCI.
Collapse
Affiliation(s)
- Todd C. Peterson
- Restorative Neuroscience Laboratory, Deptartment of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Gail D. Anderson
- Department of Pharmacy, University of Washington, Seattle, Washington
| | - Eric D. Kantor
- Department of Pharmacy, University of Washington, Seattle, Washington
| | - Michael R. Hoane
- Restorative Neuroscience Laboratory, Deptartment of Psychology, Southern Illinois University, Carbondale, Illinois
| |
Collapse
|
35
|
Scremin OU, Norman KM, Roch M, Holschneider DP, Scremin AE. Acetylcholinesterase Inhibition Interacts with Training To Reverse Spatial Learning Deficits after Cortical Impact Injury. J Neurotrauma 2012; 29:2457-64. [DOI: 10.1089/neu.2012.2465] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Oscar U. Scremin
- Research, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
- Department of Physiology, David Geffen School of Medicine at University of California–Los Angeles, Los Angeles, California
| | - Keith M. Norman
- Research, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Margareth Roch
- Research, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Daniel P. Holschneider
- Department of Psychiatry and the Behavioral Sciences, University of Southern California Keck School of Medicine, Los Angeles, California
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, California
- Department of Cell and Neurobiology, University of Southern California Keck School of Medicine, Los Angeles, California
| | - A.M. Erika Scremin
- Physical Medicine and Rehabilitation Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
- Department of Medicine, David Geffen School of Medicine at University of California–Los Angeles, Los Angeles, California
| |
Collapse
|
36
|
Choi BY, Jang BG, Kim JH, Lee BE, Sohn M, Song HK, Suh SW. Prevention of traumatic brain injury-induced neuronal death by inhibition of NADPH oxidase activation. Brain Res 2012; 1481:49-58. [PMID: 22975130 DOI: 10.1016/j.brainres.2012.08.032] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/13/2012] [Accepted: 08/16/2012] [Indexed: 11/30/2022]
Abstract
The present study aimed to evaluate the therapeutic potential of apocynin, an NADPH oxidase assembly inhibitor, on traumatic brain injury. Rat traumatic brain injury (TBI) was performed using a weight drop model. Apocynin (100mg/kg) was injected into the intraperitoneal space 15 min before TBI. Reactive oxygen species (ROS) in the hippocampal CA3 pyramidal neurons were detected by dihydroethidium (dHEt) at 3h after TBI. Oxidative injury was detected by 4-hydroxy-2-nonenal (4HNE) at 6h after TBI. Blood-brain barrier disruption was detected by IgG extravasation and neuronal death was evaluated with Fluoro Jade-B staining 24h after TBI. Microglia activation was detected by CD11b immunohistochemistry in the hippocampus at 1 week after TBI. ROS production was inhibited by apocynin administration in the hippocampal CA3 pyramidal neurons. This pre-treatment with apocynin decreased the blood-brain barrier disruption, the number of degenerating neurons in the hippocampal CA3 region and microglial activation after TBI. The present study indicates that apocynin pre-treatment prevents TBI-induced ROS production, thus decreasing BBB disruption, neuronal death and microglial activation. Therefore, the present study suggests that inhibition of NADPH oxidase by apocynin may have a high therapeutic potential to reduce traumatic brain injury-induced neuronal death.
Collapse
Affiliation(s)
- Bo Yong Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon, South Korea
| | | | | | | | | | | | | |
Collapse
|
37
|
Blaylock RL, Maroon J. Natural plant products and extracts that reduce immunoexcitotoxicity-associated neurodegeneration and promote repair within the central nervous system. Surg Neurol Int 2012; 3:19. [PMID: 22439110 PMCID: PMC3307240 DOI: 10.4103/2152-7806.92935] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 01/11/2012] [Indexed: 12/14/2022] Open
Abstract
Our understanding of the pathophysiological and biochemical basis of a number of neurological disorders has increased enormously over the last three decades. Parallel with this growth of knowledge has been a clearer understanding of the mechanism by which a number of naturally occurring plant extracts, as well as whole plants, can affect these mechanisms so as to offer protection against injury and promote healing of neurological tissues. Curcumin, quercetin, green tea catechins, balcalein, and luteolin have been extensively studied, and they demonstrate important effects on cell signaling that go far beyond their antioxidant effects. Of particular interest is the effect of these compounds on immunoexcitotoxicity, which, the authors suggest, is a common mechanism in a number of neurological disorders. By suppressing or affecting microglial activation states as well as the excitotoxic cascade and inflammatory mediators, these compounds dramatically affect the pathophysiology of central nervous system disorders and promote the release and generation of neurotrophic factors essential for central nervous system healing. We discuss the various aspects of these processes and suggest future directions for study.
Collapse
Affiliation(s)
- Russell L Blaylock
- Theoretical Neurosciences, Department of Biology, Belhaven University, Jackson, MS 39157, USA
| | | |
Collapse
|
38
|
Ullah N, Lee HY, Naseer MI, Ullah I, Suh JW, Kim MO. Nicotinamide inhibits alkylating agent-induced apoptotic neurodegeneration in the developing rat brain. PLoS One 2011; 6:e27093. [PMID: 22164206 PMCID: PMC3229474 DOI: 10.1371/journal.pone.0027093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 10/10/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Exposure to the chemotherapeutic alkylating agent thiotepa during brain development leads to neurological complications arising from neurodegeneration and irreversible damage to the developing central nerve system (CNS). Administration of single dose of thiotepa in 7-d postnatal (P7) rat triggers activation of apoptotic cascade and widespread neuronal death. The present study was aimed to elucidate whether nicotinamide may prevent thiotepa-induced neurodegeneration in the developing rat brain. METHODOLOGY/PRINCIPAL FINDINGS Neuronal cell death induced by thiotepa was associated with the induction of Bax, release of cytochrome-c from mitochondria into the cytosol, activation of caspase-3 and cleavage of poly (ADP-ribose) polymerase (PARP-1). Post-treatment of developing rats with nicotinamide suppressed thiotepa-induced upregulation of Bax, reduced cytochrome-c release into the cytosol and reduced expression of activated caspase-3 and cleavage of PARP-1. Cresyl violet staining showed numerous dead cells in the cortex hippocampus and thalamus; post-treatment with nicotinamide reduced the number of dead cells in these brain regions. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end-labeling (TUNEL) and immunohistochemical analysis of caspase-3 show that thiotepa-induced cell death is apoptotic and that it is inhibited by nicotinamide treatment. CONCLUSION Nicotinamide (Nic) treatment with thiotepa significantly improved neuronal survival and alleviated neuronal cell death in the developing rat. These data demonstrate that nicotinamide shows promise as a therapeutic and neuroprotective agent for the treatment of neurodegenerative disorders in newborns and infants.
Collapse
Affiliation(s)
- Najeeb Ullah
- Division of Life Science, College of Natural Sciences (RINS) and Applied Life Science (Brain Korea 21), Gyeongsang National University, Chinju, Republic of Korea
| | - Hae Young Lee
- Division of Life Science, College of Natural Sciences (RINS) and Applied Life Science (Brain Korea 21), Gyeongsang National University, Chinju, Republic of Korea
| | - Muhammad Imran Naseer
- Division of Life Science, College of Natural Sciences (RINS) and Applied Life Science (Brain Korea 21), Gyeongsang National University, Chinju, Republic of Korea
| | - Ikram Ullah
- Division of Life Science, College of Natural Sciences (RINS) and Applied Life Science (Brain Korea 21), Gyeongsang National University, Chinju, Republic of Korea
| | - Joo Won Suh
- Division of Bioscience and Bioinformatics, Myongji University, Namdong, Yongin, Kyonggido, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science, College of Natural Sciences (RINS) and Applied Life Science (Brain Korea 21), Gyeongsang National University, Chinju, Republic of Korea
| |
Collapse
|
39
|
Koh PO. Nicotinamide attenuates the decrease of astrocytic phosphoprotein PEA-15 in focal cerebral ischemic injury. J Vet Med Sci 2011; 74:377-80. [PMID: 22067079 DOI: 10.1292/jvms.11-0392] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nicotinamide exerts neuroprotective effects against focal cerebral ischemic injury. Phosphoprotein enriched in astrocytes 15 (PEA-15) is prominently expressed in astrocytes that exert broad anti-apoptotic functions. This study investigated whether nicotinamide modulates PEA-15 and levels of two phosphorylated PEA-15 (Serine 104 and 116) in an animal model of middle cerebral artery occlusion (MCAO)-induced injury. Adult male rats were treated with vehicle or nicotinamide (500 mg/kg) 2 hr after the onset of MCAO and cerebral cortices were collected at 24 hr after MCAO. In a proteomic approach, MCAO induced decreases of PEA-15 levels, while nicotinamide treatment attenuated the injury-induced decrease in PEA-15. The results of Western blot analysis suggest that nicotinamide prevented injury-induced reduction in phospho-PEA-15 (Serine 104) and phospho-PEA-15 (Serine 116) levels. The phosphorylation of PEA-15 exerts anti-apoptotic functions, and reduction of PEA-15 phosphorylation leads to apoptotic cell death. These results suggest that nicotinamide exerts a neuroprotective effect by attenuating the injury-induced decreases of PEA-15 and phospho-PEA-15 (Ser 104 and Ser 116) proteins.
Collapse
Affiliation(s)
- Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 660–701, South Korea.
| |
Collapse
|
40
|
Vonder Haar C, Anderson GD, Hoane MR. Continuous nicotinamide administration improves behavioral recovery and reduces lesion size following bilateral frontal controlled cortical impact injury. Behav Brain Res 2011; 224:311-7. [PMID: 21704653 PMCID: PMC3159802 DOI: 10.1016/j.bbr.2011.06.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 05/16/2011] [Accepted: 06/09/2011] [Indexed: 10/18/2022]
Abstract
Previous research has demonstrated considerable preclinical efficacy of nicotinamide (NAM; vitamin B(3)) in animal models of TBI with systemic dosing at 50 and 500 mg/kg yielding improvements on sensory, motor, cognitive and histological measures. The current study aimed to utilize a more specific dosing paradigm in a clinically relevant delivery mechanism: continuously secreting subcutaneous pumps. A bilateral frontal controlled cortical impact (CCI) or sham surgery was performed and rats were treated with NAM (150 mg/kg day) or saline (1 ml/kg) pumps 30 min after CCI, continuing until seven days post-CCI. Rats were given a loading dose of NAM (50mg/kg) or saline (1 ml/kg) following pump implant. Rats received behavioral testing (bilateral tactile adhesive removal, locomotor placing task and Morris water maze) starting on day two post-CCI and were sacrificed at 31 days post-CCI and brains were stained to examine lesion size. NAM-treated rats had reductions in sensory, motor and cognitive behavioral deficits compared to vehicle-treated rats. Specifically, NAM-treated rats significantly improved on the bilateral tactile adhesive removal task, locomotor placing task and the reference memory paradigm of the Morris water maze. Lesion size was also significantly reduced in the NAM-treated group. The results from this study indicate that at the current dose, NAM produces beneficial effects on recovery from a bilateral frontal brain injury and that it may be a relevant compound to be explored in human studies.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Life Science II, MC 6502, Southern Illinois University, Carbondale, IL 62901, USA
| | | | | |
Collapse
|
41
|
Wang Y, Wei Y, Oguntayo S, Wilkins W, Arun P, Valiyaveettil M, Song J, Long JB, Nambiar MP. Tightly coupled repetitive blast-induced traumatic brain injury: development and characterization in mice. J Neurotrauma 2011; 28:2171-83. [PMID: 21770761 DOI: 10.1089/neu.2011.1990] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A mouse model of repeated blast exposure was developed using a compressed air-driven shock tube, to study the increase in severity of traumatic brain injury (bTBI) after multiple blast exposures. Isoflurane anesthetized C57BL/6J mice were exposed to 13.9, 20.6, and 25 psi single blast overpressure (BOP1) and allowed to recover for 5 days. BOP1 at 20.6 psi showed a mortality rate of 2% and this pressure was used for three repeated blast exposures (BOP3) with 1 and 30 min intervals. Overall mortality rate in BOP3 was increased to 20%. After blast exposure, righting reflex time and body-weight loss were significantly higher in BOP3 animals compared to BOP1 animals. At 4 h, brain edema was significantly increased in BOP3 animals compared to sham controls. Reactive oxygen species in the cortex were increased significantly in BOP1 and BOP3 animals. Neuropathological analysis of the cerebellum and cerebral cortex showed dense silver precipitates in BOP3 animals, indicating the presence of diffuse axonal injury. Fluoro-Jade B staining showed increased intensity in the cortex of BOP3 animals indicating neurodegeneration. Rota Rod behavioral test showed a significant decrease in performance at 10 rpm following BOP1 or BOP3 at 2 h post-blast, which gradually recovered during the 5 days. At 20 rpm, the latency to fall was significantly decreased in both BOP1 and BOP3 animals and it did not recover in the majority of the animals through 5 days of testing. These data suggest that repeated blast exposures lead to increased impairment severity in multiple neurological parameters of TBI in mice.
Collapse
Affiliation(s)
- Ying Wang
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hoane MR, Swan AA, Heck SE. The effects of a high-fat sucrose diet on functional outcome following cortical contusion injury in the rat. Behav Brain Res 2011; 223:119-24. [PMID: 21549156 PMCID: PMC3111862 DOI: 10.1016/j.bbr.2011.04.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/10/2011] [Accepted: 04/18/2011] [Indexed: 11/26/2022]
Abstract
Traumatic brain injury (TBI) is a major public health issue affecting 1.7 million Americans each year, of which approximately 50,000 are fatal. High-fat sucrose (HFS) diets are another public health issue which can lead to obesity, hypertension, and many other debilitating disorders. These two disorders combined can lead to more complicated issues. It has recently been shown that HFS diets can reduce levels of brain-derived neurotrophic factor (BDNF) leading to reductions in neuronal and behavioral plasticity. This reduction in BDNF is suspected of increasing the susceptibility of the brain to injury. To test the effects of a HFS diet on recovery of function post-TBI, male Sprague-Dawley rats were used in this study. Eight weeks prior to TBI, rats were placed on a special HFS diet (n=14) or a standard rodent diet (n=14). Following this eight-week period, rats were prepared with bilateral frontal cortical contusion injuries (CCI) or sham procedures. Beginning two days post-TBI, animals were tested on a battery of behavioral tests to assess somatosensory dysfunction and spatial memory in the Morris water maze, with a reference memory and a working memory task. Following testing, animals were sacrificed and their brains processed for lesion analysis. The HFS diet worsened performance on the bilateral tactile adhesive removal test in sham animals. Injured animals on the Standard diet had a greater improvement in somatosensory performance in the adhesive removal test and had better performance on the working memory task compared to animals on the HFS diet. The HFS diet also resulted in significantly greater loss of cortical tissue post-CCI than in the Standard diet group. This study may aid in determining how nutritional characteristics or habits interact with damage to the brain.
Collapse
Affiliation(s)
- Michael R Hoane
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University Carbondale, Carbondale, IL, USA.
| | | | | |
Collapse
|
43
|
Sung JH, Kim MO, Koh PO. Nicotinamide prevents the down-regulation of MEK/ERK/p90RSK signaling cascade in brain ischemic injury. J Vet Med Sci 2011; 74:35-41. [PMID: 21891976 DOI: 10.1292/jvms.11-0149] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nicotinamide attenuates neuronal cell death related to focal cerebral ischemic injury. This study investigated whether nicotinamide exerts a neuroprotective effect through the activation of Raf- mitogen-activated protein kinase kinase (MEK)-ERK and its downstream targets, including p90 ribosomal S6 kinase (p90RSK) and Bad. Adult male Sprague-Dawley rats were treated with nicotinamide (500 mg/kg) or vehicle 2 hr after the onset of middle cerebral artery occlusion (MCAO). Brains were collected 24 hr after MCAO. In the present study, nicotinamide significantly reduces the volume of infarct regions and decreases the number of positive cells by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining in the cerebral cortex. Nicotinamide prevents injury-induced decrease in Raf-1, MEK1/2, and ERK1/2 phosphorylation. As part of the downstream cascade, nicotinamide inhibits the injury-induced decrease in p90RSK and Bad phosphorylation. Moreover, nicotinamide prevents the injury-induced increase in cleaved caspase-3 levels. These findings suggest that nicotinamide protects neuronal cells against cerebral ischemic injury and that MEK-ERK-p90RSK cascade activation by nicotinamide contributes to these neuroprotective effects.
Collapse
Affiliation(s)
- Jin-Hee Sung
- Department of Anatomy, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | | | | |
Collapse
|
44
|
Koh PO. Nicotinamide attenuates the ischemic brain injury-induced decrease of Akt activation and Bad phosphorylation. Neurosci Lett 2011; 498:105-9. [PMID: 21596097 DOI: 10.1016/j.neulet.2011.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 04/30/2011] [Accepted: 05/02/2011] [Indexed: 10/18/2022]
Abstract
Nicotinamide protects cortical neuronal cells against cerebral ischemic injury through activation of various cytoprotective mechanisms. Here, this study confirmed the neuroprotective effects of nicotinamide in focal cerebral ischemic injury and investigated whether nicotinamide modulates a crucial survival pathway, Akt and its downstream targets. Adult male rats were treated with vehicle or nicotinamide (500 mg/kg) 2h after the onset of middle cerebral artery occlusion (MCAO). Brains were collected 24h after MCAO and infarct volumes were analyzed. Nicotinamide significantly reduced the infarct volume in the cerebral cortex. Potential activation was measured by phosphorylation of PDK1 at Ser(241), Akt at Ser(473), and Bad at Ser(136) using Western blot analysis. Nicotinamide prevented the injury-induced decrease of pPDK1, pAkt, and pBad levels. 14-3-3 levels were not different between vehicle- and nicotinamide-treated animals. However, pBad and 14-3-3 interaction levels decreased during MCAO, but were maintained in the presence of nicotinamide, compared to levels in control animals. These findings suggest that nicotinamide attenuates cell death due to focal cerebral ischemic injury and that neuroprotective effects are mediated through the Akt signaling pathway, thus enhancing neuronal survival.
Collapse
Affiliation(s)
- Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, 900 Gajwa-dong, Jinju, South Korea.
| |
Collapse
|
45
|
Manda KR, Banerjee A, Banks WA, Ercal N. Highly active antiretroviral therapy drug combination induces oxidative stress and mitochondrial dysfunction in immortalized human blood-brain barrier endothelial cells. Free Radic Biol Med 2011; 50:801-10. [PMID: 21193030 PMCID: PMC5997409 DOI: 10.1016/j.freeradbiomed.2010.12.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 12/10/2010] [Accepted: 12/17/2010] [Indexed: 10/18/2022]
Abstract
The era of highly active antiretroviral therapy (HAART) has controlled AIDS and its related disorders considerably; however, the prevalence of HIV-1-associated neurocognitive disorders has been on the rise in the post-HAART era. In view of these developments, we investigated whether a HAART drug combination of 3'-azido-2',3'-deoxythymidine (AZT) and indinavir (IDV) can alter the functionality of the blood-brain barrier (BBB) endothelial cells, thereby exacerbating this condition. The viability of hCMEC/D3 cells (in vitro model of BBB) that were exposed to these drugs was significantly reduced after 72h treatment, in a dose-dependent manner. Reactive oxygen species were highly elevated after the exposure, indicating that mechanisms that induce oxidative stress were involved. Measures of oxidative stress parameters, such as glutathione and malondialdehyde, were altered in the treated groups. Loss of mitochondrial membrane potential, as assessed by fluorescence microscopy and decreased levels of ATP, indicated that cytotoxicity was mediated through mitochondrial dysfunction. Furthermore, AZT+IDV treatment caused apoptosis in endothelial cells, as assessed by the expression of cytochrome c and procaspase-3 proteins. Pretreatment with the thiol antioxidant N-acetylcysteine amide reversed some of the pro-oxidant effects of AZT+IDV. Results from our in vitro studies indicate that the AZT+IDV combination may affect the BBB in HIV-infected individuals treated with HAART drugs.
Collapse
Affiliation(s)
- Kalyan Reddy Manda
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO, USA
| | - Atrayee Banerjee
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO, USA
| | - William A. Banks
- GRECC-VA, Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington, Seattle, WA, USA
| | - Nuran Ercal
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO, USA
- Corresponding Author Address: Department of Chemistry, Missouri University of Science and Technology, 400 West 11th Street, Rolla, MO 65409, Phone: 573-341-6950, Fax: 573-341-6033,
| |
Collapse
|
46
|
Swan AA, Chandrashekar R, Beare J, Hoane MR. Preclinical efficacy testing in middle-aged rats: nicotinamide, a novel neuroprotectant, demonstrates diminished preclinical efficacy after controlled cortical impact. J Neurotrauma 2011; 28:431-40. [PMID: 21083416 PMCID: PMC3057203 DOI: 10.1089/neu.2010.1519] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Age is a consistent predictor of poor outcome following traumatic brain injury (TBI). Although the elderly population has one of the highest rates of TBI-related hospitalization and death, few preclinical studies have attempted to model and treat TBI in the aged population. Recent studies have indicated that nicotinamide (NAM), a soluble B-group vitamin, improved functional recovery in experimental models of TBI in young animals. The purpose of the present study was to examine the preclinical efficacy of NAM in middle-aged rats. Groups of middle-aged (14-month-old) rats were assigned to NAM (500 mg/kg or 50 mg/kg) or saline alone (1 mL/kg) treatment conditions, and received unilateral cortical contusion injuries (CCI) and injections at 1 h and 24 h following injury. The animals were tested on a variety of tasks to assess vestibulomotor (tapered beam) and cognitive performance (reference and working memory in the Morris water maze), and were evaluated for lesion size, blood-brain barrier compromise, astrocytic activation, and edema formation. In summary, the preclinical efficacy of NAM as a treatment following CCI in middle-aged rats differs from that previously documented in younger rats; while treatment with 50 mg/kg NAM appeared to have no effect, the 500-mg/kg dose worsened performance in middle-aged animals. Histological indicators demonstrated more nuanced group differences, indicating that NAM may positively impact some of the cellular cascades following injury, but were not substantial enough to improve functional recovery. These findings emphasize the need to examine potential treatments for TBI utilizing non-standard populations, and may explain why so many treatments have failed in clinical trials.
Collapse
Affiliation(s)
- Alicia A Swan
- Restorative Neuroscience Laboratory, Center for Integrative Research for Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University , Carbondale, Illinois 62901, USA
| | | | | | | |
Collapse
|
47
|
Fernandes CA, Fievez L, Ucakar B, Neyrinck AM, Fillee C, Huaux F, Delzenne NM, Bureau F, Vanbever R. Nicotinamide enhances apoptosis of G(M)-CSF-treated neutrophils and attenuates endotoxin-induced airway inflammation in mice. Am J Physiol Lung Cell Mol Physiol 2011; 300:L354-61. [DOI: 10.1152/ajplung.00198.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neutrophils constitute the first line of host defense against invading microorganisms. Yet their removal from the inflammatory environment is fundamental for injury restraint and resolution of inflammation. Nicotinamide, a component of vitamin B3, is known to modulate cell survival. In this study, we assessed the influence of nicotinamide on neutrophil apoptosis, both in vitro and in vivo in a mouse model of endotoxin-induced lung inflammation. In vitro, nicotinamide promoted apoptosis of human blood neutrophils in a dose-dependent manner in the presence of the apoptosis inhibitors granulocyte colony-stimulating factor and granulocyte/macrophage colony-stimulating factor. The highest concentration of nicotinamide completely neutralized the pro-survival effect of granulocyte (macrophage) colony-stimulating factor. Nicotinamide proapoptotic effect was associated with enhanced caspase-3 activity. In addition, nicotinamide slightly reduced neutrophil chemotaxis in vitro. In vivo, pulmonary nicotinamide delivery decreased the levels of cellular and biochemical inflammation markers and increased the percentage of apoptotic neutrophils in bronchoalveolar lavages. Our findings suggest that nicotinamide is an apoptotic stimulus for neutrophils, thereby contributing to the resolution of neutrophilic inflammation in the lungs.
Collapse
Affiliation(s)
- Cláudia A. Fernandes
- Unité de Pharmacie Galénique, Louvain Drug Research Institute, Université catholique de Louvain, Brussels
| | - Laurence Fievez
- Laboratory of Cellular and Molecular Physiology, GIGA-Research, University of Liège, Liège
| | - Bernard Ucakar
- Unité de Pharmacie Galénique, Louvain Drug Research Institute, Université catholique de Louvain, Brussels
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain,
| | - Catherine Fillee
- Department of Clinical Biology, Cliniques Universitaires Saint Luc, and
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain,
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Physiology, GIGA-Research, University of Liège, Liège
| | - Rita Vanbever
- Unité de Pharmacie Galénique, Louvain Drug Research Institute, Université catholique de Louvain, Brussels
| |
Collapse
|
48
|
Abstract
Globally, developed nations spend a significant amount of their resources on health care initiatives that poorly translate into increased population life expectancy. As an example, the United States devotes 16% of its gross domestic product to health care, the highest level in the world, but falls behind other nations that enjoy greater individual life expectancy. These observations point to the need for pioneering avenues of drug discovery to increase life span with controlled costs. In particular, innovative drug development for metabolic disorders such as diabetes mellitus becomes increasingly critical given that the number of diabetic people will increase exponentially over the next 20 years. This article discusses the elucidation and targeting of novel cellular pathways that are intimately tied to oxidative stress in diabetes mellitus for new treatment strategies. Pathways that involve wingless, β-nicotinamide adenine dinucleotide (NAD(+)) precursors, and cytokines govern complex biological pathways that determine both cell survival and longevity during diabetes mellitus and its complications. Furthermore, the role of these entities as biomarkers for disease can further enhance their utility irrespective of their treatment potential. Greater understanding of the intricacies of these unique cellular mechanisms will shape future drug discovery for diabetes mellitus to provide focused clinical care with limited or absent long-term complications.
Collapse
Affiliation(s)
- Kenneth Maiese
- Department of Neurology, 8C-1 UHC, Wayne State University School of Medicine, 4201 St. Antoine, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
49
|
Chen JF, Chern Y. Impacts of methylxanthines and adenosine receptors on neurodegeneration: human and experimental studies. Handb Exp Pharmacol 2011:267-310. [PMID: 20859800 DOI: 10.1007/978-3-642-13443-2_10] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Neurodegenerative disorders are some of the most feared illnesses in modern society, with no effective treatments to slow or halt this neurodegeneration. Several decades after the earliest attempt to treat Parkinson's disease using caffeine, tremendous amounts of information regarding the potential beneficial effect of caffeine as well as adenosine drugs on major neurodegenerative disorders have accumulated. In the first part of this review, we provide general background on the adenosine receptor signaling systems by which caffeine and methylxanthine modulate brain activity and their role in relationship to the development and treatment of neurodegenerative disorders. The demonstration of close interaction between adenosine receptor and other G protein coupled receptors and accessory proteins might offer distinct pharmacological properties from adenosine receptor monomers. This is followed by an outline of the major mechanism underlying neuroprotection against neurodegeneration offered by caffeine and adenosine receptor agents. In the second part, we discuss the current understanding of caffeine/methylxantheine and its major target adenosine receptors in development of individual neurodegenerative disorders, including stroke, traumatic brain injury Alzheimer's disease, Parkinson's disease, Huntington's disease and multiple sclerosis. The exciting findings to date include the specific in vivo functions of adenosine receptors revealed by genetic mouse models, the demonstration of a broad spectrum of neuroprotection by chronic treatment of caffeine and adenosine receptor ligands in animal models of neurodegenerative disorders, the encouraging development of several A(2A) receptor selective antagonists which are now in advanced clinical phase III trials for Parkinson's disease. Importantly, increasing body of the human and experimental studies reveals encouraging evidence that regular human consumption of caffeine in fact may have several beneficial effects on neurodegenerative disorders, from motor stimulation to cognitive enhancement to potential neuroprotection. Thus, with regard to neurodegenerative disorders, these potential benefits of methylxanthines, caffeine in particular, strongly argue against the common practice by clinicians to discourage regular human consumption of caffeine in aging populations.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
| | | |
Collapse
|
50
|
Readnower RD, Chavko M, Adeeb S, Conroy MD, Pauly JR, McCarron RM, Sullivan PG. Increase in blood-brain barrier permeability, oxidative stress, and activated microglia in a rat model of blast-induced traumatic brain injury. J Neurosci Res 2010; 88:3530-9. [PMID: 20882564 PMCID: PMC2965798 DOI: 10.1002/jnr.22510] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 07/22/2010] [Accepted: 08/06/2010] [Indexed: 11/06/2022]
Abstract
Traumatic brain injury (TBI) as a consequence of exposure to blast is increasingly prevalent in military populations, with the underlying pathophysiological mechanisms mostly unknown. In the present study, we utilized an air-driven shock tube to investigate the effects of blast exposure (120 kPa) on rat brains. Immediately following exposure to blast, neurological function was reduced. BBB permeability was measured using IgG antibody and evaluating its immunoreactivity in the brain. At 3 and 24 hr postexposure, there was a transient significant increase in IgG staining in the cortex. At 3 days postexposure, IgG immunoreactivity returned to control levels. Quantitative immunostaining was employed to determine the temporal course of brain oxidative stress following exposure to blast. Levels of 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT) were significantly increased at 3 hr postexposure and returned to control levels at 24 hr postexposure. The response of microglia to blast exposure was determined by autoradiographic localization of (3) H-PK11195 binding. At 5 days postexposure, increased binding was observed in the contralateral and ipsilateral dentate gyrus. These regions also displayed increased binding at 10 days postexposure; in addition to these regions there was increased binding in the contralateral ventral hippocampus and substantia nigra at this time point. By using antibodies against CD11b/c, microglia morphology characteristic of activated microglia was observed in the hippocampus and substantia nigra of animals exposed to blast. These results indicate that BBB breakdown, oxidative stress, and microglia activation likely play a role in the neuropathology associated with TBI as a result of blast exposure.
Collapse
Affiliation(s)
- Ryan D Readnower
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | |
Collapse
|