1
|
Mohamed-Fathy Kamal O, Ojeda-Hernández DD, Selma-Calvo B, Benito-Martín MS, Fuente-Martín SDL, García-Martín M, Larriba-González T, Sancho-Bielsa F, Matias-Guiu JA, Matias-Guiu J, Gómez-Pinedo U. Technical Assessment of Motor and Behavioral Tests in Rodent Models of Multiple Sclerosis. J Integr Neurosci 2025; 24:26429. [PMID: 40018775 DOI: 10.31083/jin26429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a neurodegenerative disorder characterized by progressive motor and cognitive impairments, affecting millions worldwide. It significantly reduces patients' quality of life and imposes a burden on health systems. Despite advances in understanding MS, there is no cure, highlighting the need for effective therapeutic strategies. Preclinical animal models are critical for gaining insights into MS pathophysiology and treatments. However, these models fail to fully replicate the complexity of human MS, making it essential to choose appropriate models and behavioral tests to evaluate their efficacy. PURPOSE This review examines various motor and cognitive behavioral tests used in preclinical MS models, discussing their strengths and limitations. The goal is to guide researchers in selecting the most appropriate tests for their models, while providing insights into how these tests are performed and analyzed. METHODS We reviewed motor and cognitive behavioral tests used in MS models, detailing test procedures and evaluating their advantages and disadvantages. RESULTS This review offers a comprehensive overview that aids researchers in choosing the most suitable tests for their studies, improving the accuracy and reliability of preclinical MS research. CONCLUSIONS Understanding the strengths and limitations of these tests is crucial for making informed decisions, leading to better experimental designs and, ultimately, more effective therapeutic interventions for MS.
Collapse
Affiliation(s)
- Ola Mohamed-Fathy Kamal
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Doddy Denise Ojeda-Hernández
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Belén Selma-Calvo
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - María Soledad Benito-Martín
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Sarah de la Fuente-Martín
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Marina García-Martín
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Teresa Larriba-González
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Francisco Sancho-Bielsa
- Área de Fisiología, Departamento de Ciencias Médicas, Facultad de Medicina de Ciudad Real, UCLM, 13071 Ciudad Real, Spain
| | - Jordi A Matias-Guiu
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
- Servicio de Neurología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Jorge Matias-Guiu
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
- Servicio de Neurología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ulises Gómez-Pinedo
- Laboratorio de Neurobiología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| |
Collapse
|
2
|
Krizman J, Colegrove D, Cunningham J, Bonacina S, Nicol T, Nerrie M, Kraus N. Concussion acutely disrupts auditory processing in division I football student-athletes. Brain Inj 2025; 39:17-25. [PMID: 39224977 PMCID: PMC11668622 DOI: 10.1080/02699052.2024.2396012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Diagnosis, assessment, and management of sports-related concussion require a multi-modal approach. Yet, currently, an objective assessment of auditory processing is not included. The auditory system is uniquely complex, relying on exquisite temporal precision to integrate signals across many synapses, connected by long axons. Given this complexity and precision, together with the fact that axons are highly susceptible to damage from mechanical force, we hypothesize that auditory processing is susceptible to concussive injury. METHODS We measured the frequency-following response (FFR), a scalp-recorded evoked potential that assesses processing of complex sound features, including pitch and phonetic identity. FFRs were obtained on male Division I Collegiate football players prior to contact practice to determine a pre-season baseline of auditory processing abilities, and again after sustaining a sports-related concussion. We predicted that concussion would decrease pitch and phonetic processing relative to the student-athlete's preseason baseline. RESULTS We found that pitch and phonetic encoding was smaller post-concussion. Student-athletes who sustained a second concussion showed similar declines after each injury. CONCLUSIONS Auditory processing should be included in the multimodal assessment of sports-related concussion. Future studies that extend this work to other sports, other injuries (e.g. blast exposure), and to female athletes are needed.
Collapse
Affiliation(s)
- Jennifer Krizman
- Department of Communication Sciences, Auditory Neuroscience Laboratory, Northwestern University, Evanston, IL, USA
| | - Danielle Colegrove
- Department of Sports Medicine, Northwestern University, Evanston, IL, USA
| | - Jenna Cunningham
- Department of Communication Sciences, Auditory Neuroscience Laboratory, Northwestern University, Evanston, IL, USA
| | - Silvia Bonacina
- Department of Communication Sciences, Auditory Neuroscience Laboratory, Northwestern University, Evanston, IL, USA
| | - Trent Nicol
- Department of Communication Sciences, Auditory Neuroscience Laboratory, Northwestern University, Evanston, IL, USA
| | - Matt Nerrie
- Department of Sports Medicine, Northwestern University, Evanston, IL, USA
| | - Nina Kraus
- Department of Communication Sciences, Auditory Neuroscience Laboratory, Northwestern University, Evanston, IL, USA
- Institute for Neuroscience, Northwestern University, Evanston, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Otolaryngology, Northwestern University, Chicago, IL, USA
- Department of Linguistics, Northwestern University, Evanston, IL, USA
| |
Collapse
|
3
|
Siedhoff HR, Chen S, Balderrama A, Jackson D, Li R, Sun GY, DePalma RG, Cui J, Gu Z. Low-Intensity Blast Exposure Induces Multifaceted Long-Lasting Anxiety-Related Behaviors in Mice. Neurotrauma Rep 2024; 5:1195-1204. [PMID: 39744609 PMCID: PMC11685500 DOI: 10.1089/neur.2024.0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
Primary blast exposure is a predominant cause of mild traumatic brain injury (mTBI) among veterans and active-duty military personnel, and affected individuals may develop long-lasting behavioral disturbances that interfere with quality of life. Our prior research with the "Missouri Blast" model demonstrated behavioral changes relevant to deficits in cognitive and affective domains after exposure to low-intensity blast (LIB). In this study, behavioral evaluations were extended to 3 months post-LIB injury using multifaceted conventional and advanced behavioral paradigms. C57BL/6J male mice, aged 2 months old, were subjected to a non-inertial primary LIB-induced mTBI by detonating 350 g of C-4 at a 3-m distance on 1-m-tall platforms. Three months after injury, mice were evaluated using the open-field test (OFT), social interaction test, and advanced Erasmus Ladder paradigm. With OFT, no apparent anxiety-like changes were detected with the LIB-exposed mice and sham controls, and both groups displayed similar center-zone activities. Although no social interaction parameters reached significance, a majority of LIB-exposed mice initiated less than 50% of interactions compared with their interaction partners, suggesting decreased sociability. With the Erasmus Ladder test to assess motor functions, associative learning, and stimulus response, LIB-exposed mice appeared to display increased instances of leaving before the cue, reminiscent of "escape behavior," indicative of anxiety-related activity different from that OFT detected. Overall, these results revealed subtle multifaceted long-lasting anxiety-relevant effects following LIB exposure. The "Missouri Blast" platform offers a basis for future research to investigate the underlying biological mechanism(s) leading to domain-specific behavioral changes.
Collapse
Affiliation(s)
- Heather R. Siedhoff
- Truman VA Hospital Research Service, Columbia, Missouri, USA
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Shanyan Chen
- Truman VA Hospital Research Service, Columbia, Missouri, USA
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Ashley Balderrama
- Truman VA Hospital Research Service, Columbia, Missouri, USA
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Dejun Jackson
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Runting Li
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Grace Y. Sun
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Ralph G. DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington, District of Columbia, USA
- Department of Surgery Uniformed Services, University of the Health Sciences, Bethesda, Maryland, USA
| | - Jiankun Cui
- Truman VA Hospital Research Service, Columbia, Missouri, USA
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Zezong Gu
- Truman VA Hospital Research Service, Columbia, Missouri, USA
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, USA
| |
Collapse
|
4
|
Maffioli E, Nonnis S, Negri A, Fontana M, Frabetti F, Rossi AR, Tedeschi G, Toni M. Environmental Temperature Variation Affects Brain Lipid Composition in Adult Zebrafish ( Danio rerio). Int J Mol Sci 2024; 25:9629. [PMID: 39273578 PMCID: PMC11394874 DOI: 10.3390/ijms25179629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
This study delves deeper into the impact of environmental temperature variations on the nervous system in teleost fish. Previous research has demonstrated that exposing adult zebrafish (Danio rerio) to 18 °C and 34 °C for 4 or 21 days induces behavioural changes compared to fish kept at a control temperature of 26 °C, suggesting alterations in the nervous system. Subsequent studies revealed that these temperature conditions also modify brain protein expression, indicating potential neurotoxic effects. The primary aim of this work was to investigate the effects of prolonged exposure (21 days) to 18 °C or 34 °C on the brain lipidomes of adult zebrafish compared to a control temperature. Analysis of the brain lipidome highlighted significant alteration in the relative abundances of specific lipid molecules at 18 °C and 34 °C, confirming distinct effects induced by both tested temperatures. Exposure to 18 °C resulted in an increase in levels of phospholipids, such as phosphatidylethanolamine, alongside a general reduction in levels of sphingolipids, including sphingomyelin. Conversely, exposure to 34 °C produced more pronounced effects, with increases in levels of phosphatidylethanolamine and those of various sphingolipids such as ceramide, gangliosides, and sphingomyelin, alongside a reduction in levels of ether phospholipids, including lysophosphatidylethanolamine ether, phosphatidylethanolamine ether, and phosphatidylglycerol ether, as well as levels of glycolipids like monogalactosyldiacylglycerol. These results, when integrated with existing proteomic and behavioural data, offer new insights into the effects of thermal variations on the nervous system in teleost fish. Specifically, our proteomic and lipidomic findings suggest that elevated temperatures may disrupt mitochondrial function, increase neuronal susceptibility to oxidative stress and cytotoxicity, alter axonal myelination, impair nerve impulse transmission, hinder synapse function and neurotransmitter release, and potentially lead to increased neuronal death. These findings are particularly relevant in the fields of cell biology, neurobiology, and ecotoxicology, especially in the context of global warming.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy
- CRC "Innovation for Well-Being and Environment" (I-WE), Università degli Studi di Milano, 20126 Milano, Italy
| | - Armando Negri
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy
| | - Manuela Fontana
- Unitech OMICs, Università degli Studi di Milano, 20139 Milan, Italy
| | - Flavia Frabetti
- Department of Medical and Surgical Sciences-DIMEC, University of Bologna, 40126 Bologna, Italy
| | - Anna Rita Rossi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy
- CRC "Innovation for Well-Being and Environment" (I-WE), Università degli Studi di Milano, 20126 Milano, Italy
| | - Mattia Toni
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy
| |
Collapse
|
5
|
Zuckerman A, Siedhoff HR, Balderrama A, Li R, Sun GY, Cifu DX, Cernak I, Cui J, Gu Z. Individualized high-resolution analysis to categorize diverse learning and memory deficits in tau rTg4510 mice exposed to low-intensity blast. Front Cell Neurosci 2024; 18:1397046. [PMID: 38948027 PMCID: PMC11212475 DOI: 10.3389/fncel.2024.1397046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/09/2024] [Indexed: 07/02/2024] Open
Abstract
Mild traumatic brain injury (mTBI) resulting from low-intensity blast (LIB) exposure in military and civilian individuals is linked to enduring behavioral and cognitive abnormalities. These injuries can serve as confounding risk factors for the development of neurodegenerative disorders, including Alzheimer's disease-related dementias (ADRD). Recent animal studies have demonstrated LIB-induced brain damage at the molecular and nanoscale levels. Nevertheless, the mechanisms linking these damages to cognitive abnormalities are unresolved. Challenges preventing the translation of preclinical studies into meaningful findings in "real-world clinics" encompass the heterogeneity observed between different species and strains, variable time durations of the tests, quantification of dosing effects and differing approaches to data analysis. Moreover, while behavioral tests in most pre-clinical studies are conducted at the group level, clinical tests are predominantly assessed on an individual basis. In this investigation, we advanced a high-resolution and sensitive method utilizing the CognitionWall test system and applying reversal learning data to the Boltzmann fitting curves. A flow chart was developed that enable categorizing individual mouse to different levels of learning deficits and patterns. In this study, rTg4510 mice, which represent a neuropathology model due to elevated levels of tau P301L, together with the non-carrier genotype were exposed to LIB. Results revealed distinct and intricate patterns of learning deficits and patterns within each group and in relation to blast exposure. With the current findings, it is possible to establish connections between mice with specific cognitive deficits to molecular changes. This approach can enhance the translational value of preclinical findings and also allow for future development of a precision clinical treatment plan for ameliorating neurologic damage of individuals with mTBI.
Collapse
Affiliation(s)
- Amitai Zuckerman
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital Research Service, Columbia, MO, United States
| | - Heather R. Siedhoff
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital Research Service, Columbia, MO, United States
| | - Ashley Balderrama
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital Research Service, Columbia, MO, United States
| | - Runting Li
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital Research Service, Columbia, MO, United States
| | - Grace Y. Sun
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - David X. Cifu
- Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Ibolja Cernak
- Thomas F. Frist, Jr. College of Medicine, Belmont University, Nashville, TN, United States
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital Research Service, Columbia, MO, United States
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital Research Service, Columbia, MO, United States
| |
Collapse
|
6
|
Yan D, Shi Y, Nan C, Jin Q, Zhuo Y, Huo H, Kong S, Zhao Z. Exosomes derived from human umbilical cord mesenchymal stem cells pretreated by monosialoteterahexosyl ganglioside alleviate intracerebral hemorrhage by down-regulating autophagy. Exp Cell Res 2024; 436:113960. [PMID: 38311048 DOI: 10.1016/j.yexcr.2024.113960] [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: 11/22/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/06/2024]
Abstract
PURPOSE Intracerebral hemorrhage (ICH) results in substantial morbidity, mortality, and disability. Depleting neural cells in advanced stages of ICH poses a significant challenge to recovery. The objective of our research is to investigate the potential advantages and underlying mechanism of exosomes obtained from human umbilical cord mesenchymal stem cells (hUMSCs) pretreated with monosialoteterahexosyl ganglioside (GM1) in the prevention of secondary brain injury (SBI) resulting from ICH. PATIENTS AND METHODS In vitro, hUMSCs were cultured and induced to differentiate into neuron-like cells after they were pretreated with 150 μg/mL GM1. The exosomes extracted from the culture medium following a 6-h pretreatment with 150 μg/mL GM1 were used as the treatment group. Striatal infusion of collagenase and hemoglobin (Hemin) was used to establish in vivo and in vitro models of ICH. RESULTS After being exposed to 150 μg/mL GM1 for 6 h, specific cells displayed typical neuron-like cell morphology and expressed neuron-specific enolase (NSE). The rate of differentiation into neuron-like cells was up to (15.9 ± 5.8) %, and the synthesis of N-Acetylgalactosaminyltransferase (GalNAcT), which is upstream of GM1, was detected by Western blot. This study presented an increase in the synthesis of GalNAcT. Compared with the ICH group, apoptosis in the treatment group was remarkably reduced, as detected by TUNEL, and mitochondrial membrane potential was restored by JC-1. Additionally, Western blot revealed the restoration of up-regulated autophagy markers Beclin-1 and LC3 and the down-regulation of autophagy marker p62 after ICH. CONCLUSION These findings suggest that GM1 is an effective agent to induce the differentiation of hUMSCs into neuron-like cells. GM1 can potentially increase GalNAcT production through "positive feedback", which generates more GM1 and promotes the differentiation of hUMSCs. After pretreatment with GM1, exosomes derived from hUMSCs (hUMSCs-Exos) demonstrate a neuroprotective effect by inhibiting autophagy in the ICH model. This study reveals the potential mechanism by which GM1 induces differentiation of hUMSCs into neuron-like cells and confirms the therapeutic effect of hUMSCs-Exos pretreated by GM1 (GM1-Exos) on an ICH model, potentially offering a new direction for stem cell therapy in ICH.
Collapse
Affiliation(s)
- Dongdong Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yunpeng Shi
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qianxu Jin
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yayu Zhuo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haoran Huo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiqi Kong
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Tentu PM, Bazaz MR, Pasam T, Shaikh AS, Rahman Z, Mourya A, Kaki VR, Madan J, Dandekar MP. Oxyberberine an oxoderivative of berberine confers neuroprotective effects in controlled-cortical impact mouse model of traumatic brain injury. Int J Neurosci 2023:1-16. [PMID: 37982448 DOI: 10.1080/00207454.2023.2286209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is known as a silent epidemic that causes many deaths and disabilities worldwide. We examined the response of oxyberberine (OBB) in lipopolysaccharide-stimulated BV2 microglial cells and a controlled-cortical impact (CCI) mouse model of TBI. METHODS We synthesized OBB from berberine, and also prepared OBB-nanocrystals (OBB-NC). Male C57BL/6 mice were used for CCI surgery, and post-CCI neurobehavioral deficits were assessed from 1 h after injury through 21 days post-injury (dpi). RESULTS OBB treatment reduced the lipopolysaccharide-triggered elevated levels of reactive oxygen species, nitric oxide, and nuclear factor kappa B (NF-κB) in BV2 microglial cells, indicating a neuroprotective potential. CCI-operated mice exhibited significant neurological deficits on 1, 3, and 5 dpi in neurological severity scoring and rotarod assay. OBB (25 and 50 mg/kg/day) and OBB-NC (3 mg/kg/day) ameliorated these neurological aberrations. Mice subjected to CCI surgery also displayed anxiogenic- and depression-like behaviours, and cognitive impairments in forced-swimming test and elevated-zero maze, and novel object recognition task, respectively. Administration of OBB reduced these long-term neuropsychiatric complications, and also levels of toll-like receptor 4 (TLR4), high-motility group protein 1 (HMGB1), NF-κB, tumour necrosis factor-alpha and interleukin 6 cytokines in the ipsilateral cortex of mice. CONCLUSION We suggest that the administration of OBB offers neuroprotective effects via inhibition of HMGB1-mediated TLR4/NFκB pathway.
Collapse
Affiliation(s)
- Priya Mounika Tentu
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Mohd Rabi Bazaz
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Tulasi Pasam
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Arbaz Sujat Shaikh
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Ziaur Rahman
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Atul Mourya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Venkata Rao Kaki
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| |
Collapse
|
8
|
Harris G, Stickland CA, Lim M, Goldberg Oppenheimer P. Raman Spectroscopy Spectral Fingerprints of Biomarkers of Traumatic Brain Injury. Cells 2023; 12:2589. [PMID: 37998324 PMCID: PMC10670390 DOI: 10.3390/cells12222589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions of people of all ages around the globe. TBI is notoriously hard to diagnose at the point of care, resulting in incorrect patient management, avoidable death and disability, long-term neurodegenerative complications, and increased costs. It is vital to develop timely, alternative diagnostics for TBI to assist triage and clinical decision-making, complementary to current techniques such as neuroimaging and cognitive assessment. These could deliver rapid, quantitative TBI detection, by obtaining information on biochemical changes from patient's biofluids. If available, this would reduce mis-triage, save healthcare providers costs (both over- and under-triage are expensive) and improve outcomes by guiding early management. Herein, we utilize Raman spectroscopy-based detection to profile a panel of 18 raw (human, animal, and synthetically derived) TBI-indicative biomarkers (N-acetyl-aspartic acid (NAA), Ganglioside, Glutathione (GSH), Neuron Specific Enolase (NSE), Glial Fibrillary Acidic Protein (GFAP), Ubiquitin C-terminal Hydrolase L1 (UCHL1), Cholesterol, D-Serine, Sphingomyelin, Sulfatides, Cardiolipin, Interleukin-6 (IL-6), S100B, Galactocerebroside, Beta-D-(+)-Glucose, Myo-Inositol, Interleukin-18 (IL-18), Neurofilament Light Chain (NFL)) and their aqueous solution. The subsequently derived unique spectral reference library, exploiting four excitation lasers of 514, 633, 785, and 830 nm, will aid the development of rapid, non-destructive, and label-free spectroscopy-based neuro-diagnostic technologies. These biomolecules, released during cellular damage, provide additional means of diagnosing TBI and assessing the severity of injury. The spectroscopic temporal profiles of the studied biofluid neuro-markers are classed according to their acute, sub-acute, and chronic temporal injury phases and we have further generated detailed peak assignment tables for each brain-specific biomolecule within each injury phase. The intensity ratios of significant peaks, yielding the combined unique spectroscopic barcode for each brain-injury marker, are compared to assess variance between lasers, with the smallest variance found for UCHL1 (σ2 = 0.000164) and the highest for sulfatide (σ2 = 0.158). Overall, this work paves the way for defining and setting the most appropriate diagnostic time window for detection following brain injury. Further rapid and specific detection of these biomarkers, from easily accessible biofluids, would not only enable the triage of TBI, predict outcomes, indicate the progress of recovery, and save healthcare providers costs, but also cement the potential of Raman-based spectroscopy as a powerful tool for neurodiagnostics.
Collapse
Affiliation(s)
- Georgia Harris
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Clarissa A. Stickland
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Matthias Lim
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pola Goldberg Oppenheimer
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Institute of Healthcare Technologies, Mindelsohn Way, Birmingham B15 2TH, UK
| |
Collapse
|
9
|
Kuang Y, Ding Q, Huang J, Yang S, Yao A, Yang X, Xiao M, Pei Q, Yang G. Pharmacokinetics, safety, and efficacy of GM1 ganglioside in healthy subjects and patients with multiple myeloma: Two dose-escalation studies. Eur J Pharm Sci 2023; 190:106565. [PMID: 37586437 DOI: 10.1016/j.ejps.2023.106565] [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: 03/21/2023] [Revised: 07/06/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
PURPOSE This study aimed to assess the pharmacokinetics, safety, and efficacy of GM1 in healthy Chinese subjects and patients with multiple myeloma. METHODS The data used in this study was derived from two dose-escalation trials: GM1-101, involving 70 healthy subjects, and GM1-201, which included 160 multiple myeloma patients. Population pharmacokinetics (PopPK) analysis was conducted on a subset of 90 participants using a nonlinear mixed-effects approach, and potential covariates were explored quantitatively. Observations of any abnormalities in vital signs, physical examinations, laboratory tests, and electrocardiograms during the study period, along with any spontaneously reported and directly observed adverse events, were documented for safety evaluation. Furthermore, neurotoxicity scales were used to assess the efficacy of GM1 as a prophylaxis for chemotherapy-induced peripheral neuropathy and to perform exposure-response analyses in conjunction with pharmacokinetic parameters. RESULTS A one-compartment model with first-order elimination best characterized the pharmacokinetics of GM1. The clearance and volume of distribution, as estimated by the final model, were 0.0942 L/h and 3.27 L for GM1-A, and 0.0714 L/h and 2.82 L for GM1-B, respectively. Covariates such as sex, body weight, and albumin significantly influenced pharmacokinetic parameters, yet the variation in steady-state exposure between subjects and reference subjects was less than 45% within their 90% confidence interval. Adverse reactions related to GM1 occurred in 20 (28.6%) and 57 (35.6%) subjects in the GM1-101 and GM1-201 cohorts, respectively. The changes in TNSc and FACT-Ntx scores from baseline at the end of periods 4 and 6 were lower in each GM1 dose group compared to the blank control group. The 400 mg dose group of GM1 displayed greater effectiveness than other dose groups. However, exposure-response analysis revealed no significant modification in efficacy with increasing GM1 exposure. CONCLUSIONS This study provides the first population pharmacokinetic analysis of GM1. GM1 exhibits a favorable safety profile among healthy subjects and patients with multiple myeloma. GM1 proved effective in mitigating chemotherapy-induced peripheral neuropathy, but this study observed no significant correlation between its efficacy and exposure. TRIAL REGISTRATION NUMBERS ChiCTR2000041283 and ChiCTR2000041283.
Collapse
Affiliation(s)
- Yun Kuang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China
| | - Qin Ding
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China
| | - Jie Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China
| | - Shuang Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China
| | - An Yao
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China
| | - Xiaoyan Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China
| | - Min Xiao
- Drug Evaluation and Adverse Drug Reaction Monitoring Center of Hunan, Changsha, China
| | - Qi Pei
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China.
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China; Department of Pharmacy, The Third Xiangya Hospital, Central South University, 172 Tongzipo Road, Changsha 410013, China; National-Local Joint Engineering Laboratory of Drug Clinical Evaluation Technology, Changsha, China.
| |
Collapse
|
10
|
Guo Z. Ganglioside GM1 and the Central Nervous System. Int J Mol Sci 2023; 24:ijms24119558. [PMID: 37298512 DOI: 10.3390/ijms24119558] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/18/2023] [Accepted: 05/04/2023] [Indexed: 06/12/2023] Open
Abstract
GM1 is one of the major glycosphingolipids (GSLs) on the cell surface in the central nervous system (CNS). Its expression level, distribution pattern, and lipid composition are dependent upon cell and tissue type, developmental stage, and disease state, which suggests a potentially broad spectrum of functions of GM1 in various neurological and neuropathological processes. The major focus of this review is the roles that GM1 plays in the development and activities of brains, such as cell differentiation, neuritogenesis, neuroregeneration, signal transducing, memory, and cognition, as well as the molecular basis and mechanisms for these functions. Overall, GM1 is protective for the CNS. Additionally, this review has also examined the relationships between GM1 and neurological disorders, such as Alzheimer's disease, Parkinson's disease, GM1 gangliosidosis, Huntington's disease, epilepsy and seizure, amyotrophic lateral sclerosis, depression, alcohol dependence, etc., and the functional roles and therapeutic applications of GM1 in these disorders. Finally, current obstacles that hinder more in-depth investigations and understanding of GM1 and the future directions in this field are discussed.
Collapse
Affiliation(s)
- Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
11
|
McQuaid C, Solorzano A, Dickerson I, Deane R. Uptake of severe acute respiratory syndrome coronavirus 2 spike protein mediated by angiotensin converting enzyme 2 and ganglioside in human cerebrovascular cells. Front Neurosci 2023; 17:1117845. [PMID: 36875642 PMCID: PMC9980911 DOI: 10.3389/fnins.2023.1117845] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction There is clinical evidence of neurological manifestations in coronavirus disease-19 (COVID-19). However, it is unclear whether differences in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/spike protein (SP) uptake by cells of the cerebrovasculature contribute to significant viral uptake to cause these symptoms. Methods Since the initial step in viral invasion is binding/uptake, we used fluorescently labeled wild type and mutant SARS-CoV-2/SP to study this process. Three cerebrovascular cell types were used (endothelial cells, pericytes, and vascular smooth muscle cells), in vitro. Results There was differential SARS-CoV-2/SP uptake by these cell types. Endothelial cells had the least uptake, which may limit SARS-CoV-2 uptake into brain from blood. Uptake was time and concentration dependent, and mediated by angiotensin converting enzyme 2 receptor (ACE2), and ganglioside (mono-sialotetrahexasylganglioside, GM1) that is predominantly expressed in the central nervous system and the cerebrovasculature. SARS-CoV-2/SPs with mutation sites, N501Y, E484K, and D614G, as seen in variants of interest, were also differentially taken up by these cell types. There was greater uptake compared to that of the wild type SARS-CoV-2/SP, but neutralization with anti-ACE2 or anti-GM1 antibodies was less effective. Conclusion The data suggested that in addition to ACE2, gangliosides are also an important entry point of SARS-CoV-2/SP into these cells. Since SARS-CoV-2/SP binding/uptake is the initial step in the viral penetration into cells, a longer exposure and higher titer are required for significant uptake into the normal brain. Gangliosides, including GM1, could be an additional potential SARS-CoV-2 and therapeutic target at the cerebrovasculature.
Collapse
Affiliation(s)
| | | | | | - Rashid Deane
- Department of Neuroscience, Del Monte Institute Neuroscience, University of Rochester, University of Rochester Medical Center (URMC), Rochester, NY, United States
| |
Collapse
|
12
|
Liu F, Simpson AB, D'Costa E, Bunn FS, van Leeuwen SS. Sialic acid, the secret gift for the brain. Crit Rev Food Sci Nutr 2022; 63:9875-9894. [PMID: 35531941 DOI: 10.1080/10408398.2022.2072270] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The human brain grows rapidly in early life which requires adequate nutrition. Human milk provides optimal nutrition for the developing brain, and breastfeeding significantly improves the cognition development of infants. These benefits have been largely attributed to human milk oligosaccharides (HMOS), associated with sialic acid (Sia). Subsequently, sialylated HMOS present a vital source of exogenous Sia to infants. Sialic acid is a key molecule essential for proper development of gangliosides, and therefore critical in brain development and function. Recent pre-clinical studies suggest dietary supplementation with Sia or sialylated oligosaccharides enhances intelligence and cognition performance in early and later life. Furthermore, emerging evidence suggests the involvement of Sia in brain homeostasis and disbalance correlates with common pathologies such as Alzheimer's disease (AD). Therefore, this review will discuss early brain health and development and the role of Sia in this process. Additionally, studies associating breastfeeding and specific HMOS to benefits in cognitive development are critically assessed. Furthermore, the review will assess studies implying the potential role of HMOS and microbiota in brain development via the gut-brain axis. Finally, the review will summarize recent advances regarding the role of Sia in neurodegenerative disease in later life and potential roles of dietary Sia sources.
Collapse
Affiliation(s)
- Fan Liu
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna Bella Simpson
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Esmée D'Costa
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fanny Sophia Bunn
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander S van Leeuwen
- Department of Laboratory Medicine, Sector Human Nutrition and Health, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Hahnefeld L, Vogel A, Gurke R, Geisslinger G, Schäfer MKE, Tegeder I. Phosphatidylethanolamine Deficiency and Triglyceride Overload in Perilesional Cortex Contribute to Non-Goal-Directed Hyperactivity after Traumatic Brain Injury in Mice. Biomedicines 2022; 10:biomedicines10040914. [PMID: 35453664 PMCID: PMC9033131 DOI: 10.3390/biomedicines10040914] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 12/10/2022] Open
Abstract
Traumatic brain injury (TBI) is often complicated by long-lasting disabilities, including headache, fatigue, insomnia, hyperactivity, and cognitive deficits. In a previous study in mice, we showed that persistent non-goal-directed hyperactivity is a characteristic post-TBI behavior that was associated with low levels of endocannabinoids in the perilesional cortex. We now analyzed lipidome patterns in the brain and plasma in TBI versus sham mice in association with key behavioral parameters and endocannabinoids. Lipidome profiles in the plasma and subcortical ipsilateral and contralateral brain were astonishingly equal in sham and TBI mice, but the ipsilateral perilesional cortex revealed a strong increase in neutral lipids represented by 30 species of triacylglycerols (TGs) of different chain lengths and saturation. The accumulation of TG was localized predominantly to perilesional border cells as revealed by Oil Red O staining. In addition, hexosylceramides (HexCer) and phosphatidylethanolamines (PE and ether-linked PE-O) were reduced. They are precursors of gangliosides and endocannabinoids, respectively. High TG, low HexCer, and low PE/PE-O showed a linear association with non-goal-directed nighttime hyperactivity but not with the loss of avoidance memory. The analyses suggest that TG overload and HexCer and PE deficiencies contributed to behavioral dimensions of post-TBI psychopathology.
Collapse
Affiliation(s)
- Lisa Hahnefeld
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, 60590 Frankfurt, Germany; (L.H.); (A.V.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), 60596 Frankfurt, Germany
| | - Alexandra Vogel
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, 60590 Frankfurt, Germany; (L.H.); (A.V.); (R.G.); (G.G.)
| | - Robert Gurke
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, 60590 Frankfurt, Germany; (L.H.); (A.V.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), 60596 Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, 60590 Frankfurt, Germany; (L.H.); (A.V.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), 60596 Frankfurt, Germany
| | - Michael K. E. Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University, 60590 Frankfurt, Germany; (L.H.); (A.V.); (R.G.); (G.G.)
- Correspondence:
| |
Collapse
|
14
|
Zelmanovich R, Pierre K, Felisma P, Cole D, Goldman M, Lucke-Wold B. High Altitude Cerebral Edema: Improving Treatment Options. BIOLOGICS 2022; 2:81-91. [PMID: 35425940 PMCID: PMC9006955 DOI: 10.3390/biologics2010007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
High altitude illness in its most severe form can lead to high altitude cerebral edema (HACE). Current strategies have focused on prevention with graduated ascents, pharmacologic prophylaxis, and descent at first signs of symptoms. Little is understood regarding treatment with steroids and oxygenation being commonly utilized. Pre-clinical studies with turmeric derivatives have offered promise due to its anti-inflammatory and antioxidant properties, but they warrant validation clinically. Ongoing work is focused on better understanding the disease pathophysiology with an emphasis on the glymphatic system and venous outflow obstruction. This review highlights what is known regarding diagnosis, treatment, and prevention, while also introducing novel pathophysiology mechanisms warranting further investigation.
Collapse
Affiliation(s)
| | - Kevin Pierre
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Patrick Felisma
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Dwayne Cole
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Matthew Goldman
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
15
|
Sphingolipid control of cognitive functions in health and disease. Prog Lipid Res 2022; 86:101162. [DOI: 10.1016/j.plipres.2022.101162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 12/14/2022]
|
16
|
Arun P, Rossetti F, Eken O, Wilder DM, Wang Y, Long JB. Phosphorylated Neurofilament Heavy Chain in the Cerebrospinal Fluid Is a Suitable Biomarker of Acute and Chronic Blast-Induced Traumatic Brain Injury. J Neurotrauma 2021; 38:2801-2810. [PMID: 34210150 DOI: 10.1089/neu.2021.0144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) has been documented as a significant concern for both military and civilian populations in response to the increased use of improvised explosive devices. Identifying biomarkers that could aid in the proper diagnosis and assessment of both acute and chronic bTBI is in urgent need since little progress has been made towards this goal. Addressing this knowledge gap is especially important in military veterans who are receiving assessment and care often years after their last blast exposure. Neuron-specific phosphorylated neurofilament heavy chain protein (pNFH) has been successfully evaluated as a reliable biomarker of different neurological disorders, as well as brain trauma resulting from contact sports and acute stages of brain injury of different origin. In the present study, we have evaluated the utility of pNFH levels measured in the cerebrospinal fluid (CSF) as an acute and chronic biomarker of brain injury resulting from single and tightly coupled repeated blast exposures using experimental rats. The pNFH levels increased at 24 h, returned to normal levels at 1 month, but increased again at 6 months and 1 year post-blast exposures. No significant changes were observed between single and repeated blast-exposed groups. To determine whether the observed increase of pNFH in CSF corresponded with its levels in the brain, we performed fluorescence immunohistochemistry in different brain regions at the four time-points evaluated. We observed decreased pNFH levels in those brain areas at 24 h, 6 months, and 1 year. The results suggest that blast exposure causes axonal degeneration at acute and chronic stages resulting in the release of pNFH, the abundant neuronal cytoskeletal protein. Moreover, the changes in pNFH levels in the CSF negatively correlated with the neurobehavioral functions in the rats, reinforcing suggestions that CSF levels of pNFH can be a suitable biomarker of bTBI.
Collapse
Affiliation(s)
- Peethambaran Arun
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Franco Rossetti
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ondine Eken
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Donna M Wilder
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ying Wang
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Joseph B Long
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| |
Collapse
|
17
|
Li Z, Zhang H, Cao C, Qian T, Li H. Gangliosides combined with mild hypothermia provides neuroprotection in a rat model of traumatic brain injury. Neuroreport 2021; 32:1113-1121. [PMID: 34284446 DOI: 10.1097/wnr.0000000000001703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Traumatic brain injury (TBI) remains a major cause of disability and death in modern society. In this study, we explored the neuroprotection role of the combination of gangliosides (GM) and mild hypothermia (MH) and the potential effect on oxidative stress injuries in a rat model of TBI. All 50 rats were randomized to five groups: (1) NC group: undergoing surgery without hit; (2) TBI group: undergoing surgery with hit; (3) GM group: TBI treated with gangliosides; (4) MHT group: TBI treated with MH; (5) GM+MHT group: TBI treated with gangliosides and MH. Spatial learning impairments, neurological function injury, Evans Blue leakage, brain MRI and oxidative stress injuries were assessed. The protein levels of Cleaved-caspase 3 and CytC were also detected. Both GM and MHT could rescue TBI-induced spatial learning impairments, improve neurological function injury and brain edema. In addition, the combination of them has a better therapeutic effect. Through the MRI, we found that compared with the TBI group, the brain tissue edema area of GM group, MHT group, and GM+MHT group was smaller, the occupancy effect was weakened, and the midline was slightly shifted. Compared with the GM group and MHT group, these changes in the GM+MHT group were much smaller. GM combined with MH-alleviated TBI-induced oxidative stress injuries and apoptosis. Our study reveals that GM and MH potentially provide neuroprotection via the suppression of oxidative stress injuries and apoptosis after TBI in rats.
Collapse
Affiliation(s)
- Zhaolin Li
- Department of Neurosurgery, The Second Hospital of Tianjin Medical University
| | - Hongwei Zhang
- Department of Neurosurgery, The Second Hospital of Tianjin Medical University
| | - Cangzhu Cao
- Department of Neurosurgery, The Second Hospital of Tianjin Medical University
| | - Tao Qian
- Department of Neurosurgery, Hebei General Hospital, Xinhua District, Shijiazhuang, China
| | - Hong Li
- Department of Neurosurgery, The Second Hospital of Tianjin Medical University
| |
Collapse
|
18
|
Zhang R, Wang J, Huang L, Wang TJ, Huang Y, Li Z, He J, Sun C, Wang J, Chen X, Wang J. The pros and cons of motor, memory, and emotion-related behavioral tests in the mouse traumatic brain injury model. Neurol Res 2021; 44:65-89. [PMID: 34308784 DOI: 10.1080/01616412.2021.1956290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a medical emergency with high morbidity and mortality. Motor, memory, and emotion-related deficits are common symptoms following TBI, yet treatment is very limited. To develop new drugs and find new therapeutic avenues, a wide variety of TBI models have been established to mimic the heterogeneity of TBI. In this regard, along with histologic measures, behavioral functional outcomes provide valuable insight into the underlying neuropathology and guide neurorehabilitation efforts for neuropsychiatric impairment after TBI. Development, characterization, and application of behavioral tests that can assess functional neurologic deficits are essential to the development of translational therapies. This comprehensive review aims to summarize 19 common behavioral tests from three aspects (motor, memory, and emotion-related) that are associated with TBI pathology. Discussion covers the apparatus, the test steps, the evaluation indexes, data collection and analysis, animal performance and applications, advantages and disadvantages as well as precautions to eliminate bias wherever possible. We discussed recent studies on TBI-related preconditioning, biomarkers, and optimized behavioral protocols. The neuropsychologic tests employed in clinics were correlated with those used in mouse TBI models. In summary, this review provides a comprehensive, up-to-date reference for TBI researchers to choose the right neurobehavioral protocol according to the research objectives of their translational investigation.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junming Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Yinrou Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zefu Li
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxin He
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Sun
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Tucker LB, Fu AH, McCabe JT. Hippocampal-Dependent Cognitive Dysfunction following Repeated Diffuse Rotational Brain Injury in Male and Female Mice. J Neurotrauma 2021; 38:1585-1606. [PMID: 33622092 PMCID: PMC8126427 DOI: 10.1089/neu.2021.0025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cognitive dysfunction is a common, often long-term complaint following acquired traumatic brain injury (TBI). Cognitive deficits suggest dysfunction in hippocampal circuits. The goal of the studies described here is to phenotype in both male and female mice the hippocampal-dependent learning and memory deficits resulting from TBI sustained by the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) device—a model that delivers both a contact–concussion injury as well as unrestrained rotational head movement. Mice sustained either sham procedures or four injuries (0.7 J, 24-h intervals). Spatial learning and memory skills assessed in the Morris water maze (MWM) approximately 3 weeks following injuries were significantly impaired by brain injuries; however, slower swimming speeds and poor performance on visible platform trials suggest that measurement of cognitive impairment with this test is confounded by injury-induced motor and/or visual impairments. A separate experiment confirmed hippocampal-dependent cognitive deficits with trace fear conditioning (TFC), a behavioral test less dependent on motor and visual function. Male mice had greater injury-induced deficits on both the MWM and TFC tests than female mice. Pathologically, the injury was characterized by white matter damage as observed by silver staining and glial fibrillary acidic protein (astrogliosis) in the optic tracts, with milder damage seen in the corpus callosum, and fimbria and brainstem (cerebral peduncles) of some animals. No changes in the density of GABAergic parvalbumin-expressing cells in the hippocampus, amygdala, or parietal cortex were found. This experiment confirmed significant sexually dimorphic cognitive impairments following a repeated, diffuse brain injury.
Collapse
Affiliation(s)
- Laura B Tucker
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda H Fu
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Finsterwald C, Dias S, Magistretti PJ, Lengacher S. Ganglioside GM1 Targets Astrocytes to Stimulate Cerebral Energy Metabolism. Front Pharmacol 2021; 12:653842. [PMID: 33995070 PMCID: PMC8115125 DOI: 10.3389/fphar.2021.653842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/13/2021] [Indexed: 02/01/2023] Open
Abstract
Gangliosides are major constituents of the plasma membrane and are known to promote a number of physiological actions in the brain, including synaptic plasticity and neuroprotection. In particular, the ganglioside GM1 was found to have a wide range of preclinical and clinical benefits in brain diseases such as spinal cord injury, Huntington’s disease and Parkinson’s disease. However, little is known about the underlying cellular and molecular mechanisms of GM1 in the brain. In the present study, we show that GM1 exerts its actions through the promotion of glycolysis in astrocytes, which leads to glucose uptake and lactate release by these cells. In astrocytes, GM1 stimulates the expression of several genes involved in the regulation of glucose metabolism. GM1 also enhances neuronal mitochondrial activity and triggers the expression of neuroprotection genes when neurons are cultured in the presence of astrocytes. Finally, GM1 leads to a neuroprotective effect in astrocyte-neuron co-culture. Together, these data identify a previously unrecognized mechanism mediated by astrocytes by which GM1 exerts its metabolic and neuroprotective effects.
Collapse
|
21
|
Leal AF, Benincore-Flórez E, Solano-Galarza D, Garzón Jaramillo RG, Echeverri-Peña OY, Suarez DA, Alméciga-Díaz CJ, Espejo-Mojica AJ. GM2 Gangliosidoses: Clinical Features, Pathophysiological Aspects, and Current Therapies. Int J Mol Sci 2020; 21:ijms21176213. [PMID: 32867370 PMCID: PMC7503724 DOI: 10.3390/ijms21176213] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022] Open
Abstract
GM2 gangliosidoses are a group of pathologies characterized by GM2 ganglioside accumulation into the lysosome due to mutations on the genes encoding for the β-hexosaminidases subunits or the GM2 activator protein. Three GM2 gangliosidoses have been described: Tay-Sachs disease, Sandhoff disease, and the AB variant. Central nervous system dysfunction is the main characteristic of GM2 gangliosidoses patients that include neurodevelopment alterations, neuroinflammation, and neuronal apoptosis. Currently, there is not approved therapy for GM2 gangliosidoses, but different therapeutic strategies have been studied including hematopoietic stem cell transplantation, enzyme replacement therapy, substrate reduction therapy, pharmacological chaperones, and gene therapy. The blood-brain barrier represents a challenge for the development of therapeutic agents for these disorders. In this sense, alternative routes of administration (e.g., intrathecal or intracerebroventricular) have been evaluated, as well as the design of fusion peptides that allow the protein transport from the brain capillaries to the central nervous system. In this review, we outline the current knowledge about clinical and physiopathological findings of GM2 gangliosidoses, as well as the ongoing proposals to overcome some limitations of the traditional alternatives by using novel strategies such as molecular Trojan horses or advanced tools of genome editing.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Eliana Benincore-Flórez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Daniela Solano-Galarza
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Rafael Guillermo Garzón Jaramillo
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Olga Yaneth Echeverri-Peña
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Diego A. Suarez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 110231, Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Correspondence: (C.J.A.-D.); (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.)
| | - Angela Johana Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Correspondence: (C.J.A.-D.); (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.)
| |
Collapse
|
22
|
He J, Russell T, Qiu X, Hao F, Kyle M, Chin L, Zhao LR. The contribution of stem cell factor and granulocyte colony-stimulating factor in reducing neurodegeneration and promoting neurostructure network reorganization after traumatic brain injury. Brain Res 2020; 1746:147000. [PMID: 32579949 DOI: 10.1016/j.brainres.2020.147000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 01/03/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in young adults worldwide. TBI-induced long-term cognitive deficits represent a growing clinical problem. Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are involved in neuroprotection and neuronal plasticity. However, the knowledge concerning reparative efficacy of SCF + G-CSF treatment in post-acute TBI recovery remains incomplete. This study aims to determine the efficacy of SCF + G-CSF on post-acute TBI recovery in young adult mice. The controlled cortical impact model of TBI was used for inducing a severe damage in the motor cortex of the right hemisphere in 8-week-old male C57BL mice. SCF + G-CSF treatment was initiated 3 weeks after induction of TBI. Severe TBI led to persistent motor functional deficits (Rota-Rod test) and impaired spatial learning function (water maze test). SCF + G-CSF treatment significantly improved the severe TBI-impaired spatial learning function 6 weeks after treatment. TBI also caused significant increases of Fluoro-Jade C positive degenerating neurons in bilateral frontal cortex, striatum and hippocampus, and significant reductions in MAP2+ apical dendrites and overgrowth of SMI312+ axons in peri-TBI cavity frontal cortex and in the ipsilateral hippocampal CA1 at 24 weeks post-TBI. SCF + G-CSF treatment significantly reduced TBI-induced neurodegeneration in the contralateral frontal cortex and hippocampal CA1, increased MAP2+ apical dendrites in the peri-TBI cavity frontal cortex, and prevented TBI-induced axonal overgrowth in both the peri-TBI cavity frontal cortex and ipsilateral hippocampal CA1.These findings reveal a novel pathology of axonal overgrowth after severe TBI and demonstrate a therapeutic potential of SCF + G-CSF in ameliorating severe TBI-induced long-term neuronal pathology, neurostructural network malformation, and impairments in spatial learning.
Collapse
Affiliation(s)
- Junchi He
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Thomas Russell
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Xuecheng Qiu
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Fei Hao
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Michele Kyle
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Lawrence Chin
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
23
|
Zeng XJ, Li P, Ning YL, Zhao Y, Peng Y, Yang N, Xu YW, Chen JF, Zhou YG. A 2A R inhibition in alleviating spatial recognition memory impairment after TBI is associated with improvement in autophagic flux in RSC. J Cell Mol Med 2020; 24:7000-7014. [PMID: 32394486 PMCID: PMC7299719 DOI: 10.1111/jcmm.15361] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/12/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
Spatial recognition memory impairment is an important complication after traumatic brain injury (TBI). We previously found that spatial recognition memory impairment can be alleviated in adenosine A2A receptor knockout (A2AR KO) mice after TBI, but the mechanism remains unclear. In the current study, we used manganese‐enhanced magnetic resonance imaging and the Y‐maze test to determine whether the electrical activity of neurons in the retrosplenial cortex (RSC) was reduced and spatial recognition memory was impaired in wild‐type (WT) mice after moderate TBI. Furthermore, spatial recognition memory was damaged by optogenetically inhibiting the electrical activity of RSC neurons in WT mice. Additionally, the electrical activity of RSC neurons was significantly increased and spatial recognition memory impairment was reduced in A2AR KO mice after moderate TBI. Specific inhibition of A2AR in the ipsilateral RSC alleviated the impairment in spatial recognition memory in WT mice. In addition, A2AR KO improved autophagic flux in the ipsilateral RSC after injury. In primary cultured neurons, activation of A2AR reduced lysosomal‐associated membrane protein 1 and cathepsin D (CTSD) levels, increased phosphorylated protein kinase A and phosphorylated extracellular signal‐regulated kinase 2 levels, reduced transcription factor EB (TFEB) nuclear localization and impaired autophagic flux. These results suggest that the impairment of spatial recognition memory after TBI may be associated with impaired autophagic flux in the RSC and that A2AR activation may reduce lysosomal biogenesis through the PKA/ERK2/TFEB pathway to impair autophagic flux.
Collapse
Affiliation(s)
- Xu-Jia Zeng
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Li
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ya-Lei Ning
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Zhao
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Peng
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Yang
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Ya-Wei Xu
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiang-Fan Chen
- Department of Neurology and Pharmacology, Boston University School of Medicine, Boston, MA, USA
| | - Yuan-Guo Zhou
- State Key Laboratory of Trauma, Burn, and Combined Injury, Department of Occupational Disease, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
24
|
Tang W, Fang F, Liu K, Huang Z, Li H, Yin Y, Wang J, Wang G, Wei L, Ou Y, Wang Y. Aligned Biofunctional Electrospun PLGA-LysoGM1 Scaffold for Traumatic Brain Injury Repair. ACS Biomater Sci Eng 2020; 6:2209-2218. [PMID: 33455302 DOI: 10.1021/acsbiomaterials.9b01636] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Due to poor regenerative capabilities of the brain, a treatment for traumatic brain injury (TBI) presents a serious challenge to modern medicine. Biofunctional scaffolds that can support neuronal growth, guide neurite elongation, and re-establish impaired brain tissues are urgently needed. To this end, we developed an aligned biofunctional scaffold (aPLGA-LysoGM1), in which poly (lactic-co-glycolic acid) (PLGA) was functionalized with sphingolipid ceramide N-deacylase (SCDase)-hydrolyzed monosialotetrahexosylganglioside (LysoGM1) and electrospinning was used to form an aligned fibrous network. As a ganglioside of neuronal membranes, the functionalized LysoGM1 endows the scaffold with unique biological properties favoring the growth of neuron and regeneration of injured brain tissues. Moreover, we found that the aligned PLGA-LysoGM1 fibers acted as a topographical cue to guide neurite extension, which is critical for organizing the formation of synaptic networks (neural networks). Systematic in vitro studies demonstrated that the aligned biofunctional scaffold promotes neuronal viability, neurite outgrowth, and synapse formation and also protects neurons from pressure-related injury. Additionally, in a rat TBI model, we demonstrated that the implantation of aPLGA-LysoGM1 scaffold supported recovery from brain injury, as more endogenous neurons were found to migrate and infiltrate into the defect zone compared with alternative scaffold. These results suggest that the aligned biofunctional aPLGA-LysoGM1 scaffold represents a promising therapeutic strategy for brain tissue regeneration following TBI.
Collapse
Affiliation(s)
- Wei Tang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Fang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ke Liu
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhi Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Hui Li
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ying Yin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jun Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Guocheng Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liyu Wei
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yun Ou
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Hunan Provincial Key Laboratory of Health Maintenance for Mechanical Equipment, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yazhou Wang
- School of Medicine, Chongqing University, Chongqing 400044, China.,Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
25
|
Harper MM, Rudd D, Meyer KJ, Kanthasamy AG, Anantharam V, Pieper AA, Vázquez-Rosa E, Shin MK, Chaubey K, Koh Y, Evans LP, Bassuk AG, Anderson MG, Dutca L, Kudva IT, John M. Identification of chronic brain protein changes and protein targets of serum auto-antibodies after blast-mediated traumatic brain injury. Heliyon 2020; 6:e03374. [PMID: 32099918 PMCID: PMC7029173 DOI: 10.1016/j.heliyon.2020.e03374] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/19/2019] [Accepted: 02/03/2020] [Indexed: 12/13/2022] Open
Abstract
In addition to needing acute emergency management, blast-mediated traumatic brain injury (TBI) is also a chronic disorder with delayed-onset symptoms that manifest and progress over time. While the immediate consequences of acute blast injuries are readily apparent, chronic sequelae are harder to recognize. Indeed, the identification of individuals with mild-TBI or TBI-induced symptoms is greatly impaired in large part due to the lack of objective and robust biomarkers. The purpose of this study was to address these need by identifying candidates for serum-based biomarkers of blast TBI, and also to identify unique or differentially regulated protein expression in the thalamus in C57BL/6J mice exposed to blast using high throughput qualitative screens of protein expression. To identify thalamic proteins differentially or uniquely associated with blast exposure, we utilized an antibody-based affinity-capture strategy (referred to as "proteomics-based analysis of depletomes"; PAD) to deplete thalamic lysates from blast-treated mice of endogenous thalamic proteins also found in control mice. Analysis of this "depletome" detected 75 unique proteins, many with associations to the myelin sheath. To identify blast-associated proteins eliciting production of circulating autoantibodies, serum antibodies of blast-treated mice were immobilized, and their immunogens subsequently identified by proteomic analysis of proteins specifically captured following incubation with thalamic lysates (a variant of a strategy referred to as "proteomics-based expression library screening"; PELS). This analysis identified 46 blast-associated immunogenic proteins, including 6 shared in common with the PAD analysis (ALDOA, PHKB, HBA-A1, DPYSL2, SYN1, and CKB). These proteins and their autoantibodies are appropriate for further consideration as biomarkers of blast-mediated TBI.
Collapse
Affiliation(s)
- Matthew M. Harper
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, USA
- The University of Iowa Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Danielle Rudd
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, USA
| | - Kacie J. Meyer
- The University of Iowa Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | | | | | - Andrew A. Pieper
- Harrington Discovery Institute, University Hospitals of Cleveland, Department of Psychiatry Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes VA Medical Center, Cleveland, OH 44106, USA
| | - Edwin Vázquez-Rosa
- Harrington Discovery Institute, University Hospitals of Cleveland, Department of Psychiatry Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes VA Medical Center, Cleveland, OH 44106, USA
| | - Min-Kyoo Shin
- Harrington Discovery Institute, University Hospitals of Cleveland, Department of Psychiatry Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes VA Medical Center, Cleveland, OH 44106, USA
| | - Kalyani Chaubey
- Harrington Discovery Institute, University Hospitals of Cleveland, Department of Psychiatry Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes VA Medical Center, Cleveland, OH 44106, USA
| | - Yeojung Koh
- Harrington Discovery Institute, University Hospitals of Cleveland, Department of Psychiatry Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes VA Medical Center, Cleveland, OH 44106, USA
| | - Lucy P. Evans
- The University of Iowa Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- The University of Iowa Department of Neurology, University of Iowa, Iowa City, IA, USA
- The University of Iowa Department of Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Alexander G. Bassuk
- The University of Iowa Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- The University of Iowa Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - Michael G. Anderson
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, USA
- The University of Iowa Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- The University of Iowa Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Laura Dutca
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, USA
| | - Indira T. Kudva
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA
| | | |
Collapse
|
26
|
Fallon BA, Strobino B, Reim S, Stoner J, Cunningham MW. Anti-lysoganglioside and other anti-neuronal autoantibodies in post-treatment Lyme Disease and Erythema Migrans after repeat infection. Brain Behav Immun Health 2020; 2:100015. [PMID: 34589824 PMCID: PMC8474536 DOI: 10.1016/j.bbih.2019.100015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/23/2019] [Accepted: 11/23/2019] [Indexed: 02/04/2023] Open
Abstract
Background Molecular mimicry targeting neural tissue has been reported after Borrelia burgdorferi(Bb) infection. Herein, we investigate whether antineuronal autoantibodies are increased and whether antibody-mediated signaling of neuronal cells is elevated in a cohort of symptomatic adults with a history of Lyme Disease (LD). Methods Participants (n = 179) included 24 with recent Erythema Migrans (EM) without prior LD, 8 with recent EM and prior LD (EM + prior LD), 119 with persistent post-treatment LD symptoms (PTLS), and 28 seronegative endemic controls with no prior LD history. Antineuronal immunoglobulin G (IgG) titers were measured by standard ELISA and compared with mean titers of normal age-matched sera against lysoganglioside, tubulin, and dopamine receptors (D1R and D2R). Antibody-mediated signaling of calcium calmodulin dependent protein kinase II (CaMKII) activity in a human neuronal cell line (SK-N-SH) was identified in serum. Results EM + prior LD cases had higher antibody titers than controls for anti-lysoganglioside GM1 (p = 0.002), anti-tubulin (p = 0.03), and anti-D1R (p = 0.02), as well as higher expression in the functional antibody-mediated CaMKII Assay (p = 0.03). The EM cases with no prior history showed no significant differences on any measures. The PTLS cases demonstrated significantly higher titers (p = 0.01) than controls on anti-lysoganglioside GM1, but not for the other measures. Conclusion The finding of elevated anti-neuronal autoantibodies in our small sample of those with a prior history of Lyme disease but not in those without prior Lyme disease, if replicated in a larger sample, suggests an immune priming effect of repeated infection; the CaMKII activation suggests that antineuronal antibodies have functional significance. The elevation of anti-lysoganglioside antibodies among those with PTLS is of particular interest given the established role of anti-ganglioside antibodies in peripheral and central neurologic diseases. Future prospective studies can determine whether these autoantibodies emerge after Bb infection and whether their emergence coincides with persistent neurologic or neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Brian A. Fallon
- Columbia Psychiatry, Columbia University Irving Medical Center, New York, USA
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, USA
- Corresponding author. Columbia University, 1051 Riverside Drive, Unit 69, New York, NY, 10032, USA.
| | - Barbara Strobino
- Columbia Psychiatry, Columbia University Irving Medical Center, New York, USA
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, USA
| | - Sean Reim
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Julie Stoner
- Department of Biostatistics, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Madeleine W. Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| |
Collapse
|
27
|
Niu F, Qian K, Qi H, Zhao Y, Jiang Y, Jia W, Sun M. CPCGI Reduces Gray and White Matter Injury by Upregulating Nrf2 Signaling and Suppressing Calpain Overactivation in a Rat Model of Controlled Cortical Impact. Neuropsychiatr Dis Treat 2020; 16:1929-1941. [PMID: 32904488 PMCID: PMC7455756 DOI: 10.2147/ndt.s266136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/28/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Compound porcine cerebroside and ganglioside injection (CPCGI), which involves injection of a neurotrophic drug, has been widely used to treat certain brain disorders in the clinic; however, the detailed mechanism is unknown. This study investigated whether CPCGI protects the brain from trauma by stimulating antioxidative nuclear factor erythroid-2-related factor 2 (Nrf2) signaling and suppressing calpain overactivation in a rat model of controlled cortical impact (CCI). MATERIALS AND METHODS The rat model of CCI was used. Neurological deficits, contusion, and white matter damage were evaluated 3 days after CCI. Calpain activation, Nrf2 signaling and oxidative stress were determined 24 h after CCI. RESULTS CPCGI dose-dependently reduced neurological deficits, attenuated axonal and myelin sheath injury, and decreased contusion volume 3 days post-CCI. Moreover, CPCGI reduced calpain activity, and enhanced the cytosolic levels of calpastatin, αII-spectrin, microtubule-associated protein 2 (MAP2), neurofilament heavy chain (NF-H) and myelin basic protein (MBP) in traumatic tissues 24 h post-CCI. Furthermore, CPCGI reduced the levels of nuclear Kelch-like ECH-associated protein 1 (Keap1) and thioredoxin interacting protein (TXNIP); increased the levels of cytosolic Nrf2 and thioredoxin 1 (Trx 1) and nuclear Nrf2; increased the cytosolic and nuclear Nrf2/Keap1 and Trx 1/TXNIP ratios; enhanced the levels of heme oxygenase 1 (HO-1), glutathione (GSH), superoxide dismutase activity, and total antioxidative capacity; and reduced the levels of malondialdehyde in TBI tissues. CONCLUSION These data confirm the neuroprotective effect of CPCGI against gray and white matter damage due to CCI and suggest that activating Nrf2 signaling and alleviating oxidative stress-mediated calpain activation could be one mechanism by which CPCGI protects against brain trauma.
Collapse
Affiliation(s)
- Fei Niu
- Department of Neurotrauma, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Ke Qian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, People's Republic of China
| | - Hongyan Qi
- Department of Acupuncture, Lianyungang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang City 222000, Jiangsu Province, People's Republic of China
| | - Yumei Zhao
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Yingying Jiang
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, People's Republic of China
| | - Ming Sun
- Department of Neuropharmacology, Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| |
Collapse
|
28
|
Arun P, Wilder DM, Eken O, Urioste R, Batuure A, Sajja S, Van Albert S, Wang Y, Gist ID, Long JB. Long-Term Effects of Blast Exposure: A Functional Study in Rats Using an Advanced Blast Simulator. J Neurotrauma 2019; 37:647-655. [PMID: 31595810 DOI: 10.1089/neu.2019.6591] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Anecdotal observations of blast victims indicate that significant neuropathological and neurobehavioral defects may develop at later stages of life. To pre-clinically model this phenomenon, we have examined neurobehavioral changes in rats up to 1 year after exposure to single and tightly coupled repeated blasts using an advanced blast simulator. Neurobehavioral changes were monitored at acute, sub-acute, and chronic time-points using Morris water maze test of spatial learning and memory, novel object recognition test of short-term memory, open field exploratory activity as a test of anxiety/depression, a rotating pole test for vestibulomotor function, and a rotarod balance test for motor coordination. Single and repeated blasts resulted in significant functional deficits at both acute and chronic time-points. In most functional tests, rats exposed to repeated blasts performed more poorly than rats exposed to single blast. Interestingly, several functional deficits post-blast were most pronounced at 6 months and beyond. Significant neuromotor impairments occurred at early stages after blast exposure and the severity increased with repeated exposures. The novel object recognition testing revealed short-term memory deficits at 6 and 12 months post-blast. The water maze test revealed impairments at acute and chronic stages after blast exposure. The most substantial changes in the blast-exposed rats were observed with the center time and margin time legacies in the open field exploration test at 6, 9, and 12 months post-blast. Notably, these two outcome measures were minimally altered acutely, recovered during sub-acute stages, and were markedly affected during the chronic stages after blast exposures and may implicate development of chronic anxiety and depressive-like behaviors.
Collapse
Affiliation(s)
- Peethambaran Arun
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Donna M Wilder
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Ondine Eken
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Rodrigo Urioste
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Andrew Batuure
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Sujith Sajja
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Stephen Van Albert
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Ying Wang
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Irene D Gist
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Joseph B Long
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| |
Collapse
|
29
|
Magistretti PJ, Geisler FH, Schneider JS, Li PA, Fiumelli H, Sipione S. Gangliosides: Treatment Avenues in Neurodegenerative Disease. Front Neurol 2019; 10:859. [PMID: 31447771 PMCID: PMC6691137 DOI: 10.3389/fneur.2019.00859] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/24/2019] [Indexed: 01/09/2023] Open
Abstract
Gangliosides are cell membrane components, most abundantly in the central nervous system (CNS) where they exert among others neuro-protective and -restorative functions. Clinical development of ganglioside replacement therapy for several neurodegenerative diseases was impeded by the BSE crisis in Europe during the 1990s. Nowadays, gangliosides are produced bovine-free and new pre-clinical and clinical data justify a reevaluation of their therapeutic potential in neurodegenerative diseases. Clinical experience is greatest with monosialo-tetrahexosyl-ganglioside (GM1) in the treatment of stroke. Fourteen randomized controlled trials (RCTs) in overall >2,000 patients revealed no difference in survival, but consistently superior neurological outcomes vs. placebo. GM1 was shown to attenuate ischemic neuronal injuries in diabetes patients by suppression of ERK1/2 phosphorylation and reduction of stress to the endoplasmic reticulum. There is level-I evidence from 5 RCTs of a significantly faster recovery with GM1 vs. placebo in patients with acute and chronic spinal cord injury (SCI), disturbance of consciousness after subarachnoid hemorrhage, or craniocerebral injuries due to closed head trauma. In Parkinson's disease (PD), two RCTs provided evidence of GM1 to be superior to placebo in improving motor symptoms and long-term to result in a slower than expected symptom progression, suggesting disease-modifying potential. In Alzheimer's disease (AD), the role of gangliosides has been controversial, with some studies suggesting a "seeding" role for GM1 in amyloid β polymerization into toxic forms, and others more recently suggesting a rather protective role in vivo. In Huntington's disease (HD), no clinical trials have been conducted yet. However, low GM1 levels observed in HD cells were shown to increase cell susceptibility to apoptosis. Accordingly, treatment with GM1 increased survival of HD cells in vitro and consistently ameliorated pathological phenotypes in several murine HD models, with effects seen at molecular, cellular, and behavioral level. Given that in none of the clinical trials using GM1 any clinically relevant safety issues have occurred to date, current data supports expanding GM1 clinical research, particularly to conditions with high, unmet medical need.
Collapse
Affiliation(s)
- Pierre J. Magistretti
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Fred H. Geisler
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jay S. Schneider
- Parkinson's Disease Research Unit, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Technology Enterprise (BRITE), North Carolina Central University, Durham, NC, United States
| | - Hubert Fiumelli
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Simonetta Sipione
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
GM1 ganglioside prevents axonal regeneration inhibition and cognitive deficits in a mouse model of traumatic brain injury. Sci Rep 2018; 8:13340. [PMID: 30190579 PMCID: PMC6127193 DOI: 10.1038/s41598-018-31623-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/13/2018] [Indexed: 11/08/2022] Open
Abstract
Traumatic Brain Injury (TBI) is one of the most common causes of neurological damage in young populations. It has been previously suggested that one of the mechanisms that underlie brain injury is Axonal Outgrowth Inhibition (AOI) that is caused by altered composition of the gangliosides on the axon surface. In the present study, we have found a significant reduction of GM1 ganglioside levels in the cortex in a closed head traumatic brain injury model of a mouse, induced by a weight drop device. In addition, axonal regeneration in the brains of the injured mice was affected as seen by the expression of the axonal marker pNF-H and the growth cones (visualized by F-actin and β-III-tubulin). NeuN immunostaining revealed mTBI-induced damage to neuronal survival. Finally, as expected, spatial and visual memories (measured by the Y-maze and the Novel Object Recognition tests, respectively) were also damaged 7 and 30 days post injury. A single low dose of GM1 shortly after the injury (2 mg/kg; IP) prevented all of the deficits mentioned above. These results reveal additional insights into the neuroprotective characteristics of GM1 in prevention of biochemical, cellular and cognitive changes caused by trauma, and may suggest a potential intervention for mTBI.
Collapse
|
31
|
Caughlin S, Hepburn J, Liu Q, Wang L, Yeung KKC, Cechetto DF, Whitehead SN. Chloroquine Restores Ganglioside Homeostasis and Improves Pathological and Behavioral Outcomes Post-stroke in the Rat. Mol Neurobiol 2018; 56:3552-3562. [PMID: 30145786 DOI: 10.1007/s12035-018-1317-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022]
Abstract
Perturbations of ganglioside homeostasis have been observed following stroke whereby toxic simple gangliosides GM2 and GM3 accumulate, while protective complex species GM1 and GD1 are reduced. Thus, there is a need for therapeutic interventions which can prevent ganglioside dysregulation after stroke. A pharmacological intervention using chloroquine was selected for its transient lysosomotropic properties which disrupt the activity of catabolic ganglioside enzymes. Chloroquine was administered both in vitro (0.1 μM), to primary cortical neurons exposed to GM3 toxicity, and in vivo (45 mg/kg i.p.), to 3-month-old male Wistar rats that underwent a severe stroke injury. Chloroquine was administered for seven consecutive days beginning 3 days prior to the stroke injury. Gangliosides were examined using MALDI imaging mass spectrometry at 3 and 21 days after the injury, and motor deficits were examined using the ladder task. Chloroquine treatment prevented ganglioside dysregulation 3 days post-stroke and partially prevented complex ganglioside depletion 21 days post-stroke. Exogenous GM3 was found to be toxic to primary cortical neurons which was protected by chloroquine treatment. Motor deficits were prevented in the forelimbs of stroke-injured rats with chloroquine treatment and was associated with decreased inflammation, neurodegeneration, and an increase in cell survival at the site of injury. Chloroquine administration prevents ganglioside dysregulation acutely, protects against GM3 toxicity in neurons, and is associated with long-term functional and pathological improvements after stroke in the rat. Therefore, targeting lipid dysregulation using lysosomotropic agents such as chloroquine may represent a novel therapeutic avenue for stroke injuries.
Collapse
Affiliation(s)
- Sarah Caughlin
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Jeffrey Hepburn
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Qingfan Liu
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Lynn Wang
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Ken K-C Yeung
- Department of Chemistry, Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - David F Cechetto
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Shawn N Whitehead
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
32
|
Ma D, Wang N, Fan X, Zhang L, Luo Y, Huang R, Zhang L, Li Y, Zhao G, Li L. Protective Effects of Cornel Iridoid Glycoside in Rats After Traumatic Brain Injury. Neurochem Res 2018; 43:959-971. [PMID: 29492766 DOI: 10.1007/s11064-018-2501-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 02/01/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
Cornel iridoid glycoside (CIG) is the active ingredient extracted from Cornus officinalis. Our previous studies showed that CIG had protective effects on several brain injury models. In the present study, we aimed to examine the effects and elucidate the mechanisms of CIG against traumatic brain injury (TBI). TBI was induced in the right cerebral cortex of male adult rats. The neurological and cognitive functions were evaluated by modified neurological severity score (mNSS) and object recognition test (ORT), respectively. The level of serum S100β was measured by an ELISA method. Nissl staining was used to estimate the neuron survival in the brain. The expression of proteins was determined by western blot and/or immunohistochemical staining. We found that intragastric administration of CIG in TBI rats ameliorated the neurological defects and cognitive impairment, and alleviated the neuronal loss in the injured brain. In the acute stage of TBI (24-72 h), CIG decreased the level of S100β in the serum and brain, increased the ratio of Bcl-2/Bax and decreased the expression of caspase-3 in the injured cortex. Moreover, the treatment with CIG for 30 days increased the levels of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), enhanced the expression of synapsin I, synaptophysin and postsynaptic density protein 95 (PSD-95), and inhibited the apoptosis-regulating factors in the chronic stage of TBI. The present study demonstrated that CIG had neuroprotective effects against TBI through inhibiting apoptosis in the acute stage and promoting neurorestoration in the chronic stage. The results suggest that CIG may be beneficial to TBI therapy.
Collapse
Affiliation(s)
- Denglei Ma
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Na Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Xiaotong Fan
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University; Beijing Institute for Brain Disorders, Beijing, 100053, China
| | - Lan Zhang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Yi Luo
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Rui Huang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Li Zhang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Yali Li
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University; Beijing Institute for Brain Disorders, Beijing, 100053, China.
| | - Lin Li
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University; Beijing Engineering Research Center for Nerve System Drugs; Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
33
|
Gong G, Yin L, Yuan L, Sui D, Sun Y, Fu H, Chen L, Wang X. Ganglioside GM1 protects against high altitude cerebral edema in rats by suppressing the oxidative stress and inflammatory response via the PI3K/AKT-Nrf2 pathway. Mol Immunol 2018; 95:91-98. [PMID: 29428576 DOI: 10.1016/j.molimm.2018.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 02/07/2023]
Abstract
High altitude cerebral edema (HACE) is a severe type of acute mountain sickness (AMS) that occurs in response to a high altitude hypobaric hypoxic (HH) environment. GM1 monosialoganglioside can alleviate brain injury under adverse conditions including amyloid-β-peptide, ischemia and trauma. However, its role in HACE-induced brain damage remains poorly elucidated. In this study, GM1 supplementation dose-dependently attenuated increase in rat brain water content (BWC) induced by hypobaric chamber (7600 m) exposurefor 24 h. Compared with the HH-treated group, rats injected with GM1 exhibited less brain vascular leakage, lower aquaporin-4 and higher occludin expression, but they also showed increase in Na+/K+-ATPase pump activities. Importantly, HH-incurred consciousness impairment and coordination loss also were ameliorated following GM1 administration. Furthermore, the increased oxidative stress and decrease in anti-oxidant stress system under the HH condition were also reversely abrogated by GM1 treatment via suppressing accumulation of ROS, MDA and elevating the levels of SOD and GSH. Simultaneously, GM1 administration also counteracted the enhanced inflammation in HH-exposed rats by muting pro-inflammatory cytokines IL-1β, TNF-α, and IL-6 levels in serum and brain tissues. Subsequently, GM1 potentiated the activation of the PI3K/AKT-Nrf2 pathway. Cessation of this pathway by LY294002 reversed GM1-mediated inhibitory effects on oxidative stress and inflammation, and ultimately abrogated the protective role of GM1 in abating brain edema, cognitive and motor dysfunction. Overall, GM1 may afford a protective intervention in HACE by suppressing oxidative stress and inflammatory response via activating the PI3K/AKT-Nrf2 pathway, implying a promising agent for the treatment of HACE.
Collapse
Affiliation(s)
- Gu Gong
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Liang Yin
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Libang Yuan
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Daming Sui
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Yangyang Sun
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Haiyu Fu
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Liang Chen
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Xiaowu Wang
- Center of Cardiovascular Surgery, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, PR China.
| |
Collapse
|
34
|
Zhou J, Liu T, Cui H, Fan R, Zhang C, Peng W, Yang A, Zhu L, Wang Y, Tang T. Xuefu zhuyu decoction improves cognitive impairment in experimental traumatic brain injury via synaptic regulation. Oncotarget 2017; 8:72069-72081. [PMID: 29069769 PMCID: PMC5641112 DOI: 10.18632/oncotarget.18895] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/12/2017] [Indexed: 11/25/2022] Open
Abstract
An overarching consequence of traumatic brain injury (TBI) is the cognitive impairment. It may hinder individual performance of daily tasks and determine people's subjective well-being. The damage to synaptic plasticity, one of the key mechanisms of cognitive dysfunction, becomes the potential therapeutic strategy of TBI. In this study, we aimed to investigate whether Xuefu Zhuyu Decoction (XFZYD), a traditional Chinese medicine, provided a synaptic regulation to improve cognitive disorder following TBI. Morris water maze and modified neurological severity scores were performed to assess the neurological and cognitive abilities. The PubChem Compound IDs of the major compounds of XFZYD were submitted into BATMAN-TCM, an online bioinformatics analysis tool, to predict the druggable targets related to synaptic function. Furthermore, we validated the prediction through immunohistochemical, RT-PCR and western blot analyses. We found that XFZYD enhanced neuroprotection, simultaneously improved learning and memory performances in controlled cortical impact rats. Bioinformatics analysis revealed that the improvements of XFZYD implied the Long-term potentiation relative proteins including NMDAR1, CaMKII and GAP-43. The further confirmation of molecular biological studies confirmed that XFZYD upregulated the mRNA and protein levels of NMDAR1, CaMKII and GAP-43. Pharmacological synaptic regulation of XFZYD could provide a novel therapeutic strategy for cognitive impairment following TBI.
Collapse
Affiliation(s)
- Jing Zhou
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Tao Liu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
- Department of Gerontology, Traditional Chinese Medicine Hospital Affiliate to Xinjiang Medical University, 830000 Urumqi, China
| | - Hanjin Cui
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Rong Fan
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Chunhu Zhang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Weijun Peng
- Department of Traditional Chinese Medicine, 2nd Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Ali Yang
- Department of Neurology, Henan Province People’ Hospital, 450003 Zhengzhou, China
| | - Lin Zhu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Tao Tang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| |
Collapse
|
35
|
The Invisibility of Mild Traumatic Brain Injury: Impaired Cognitive Performance as a Silent Symptom. J Neurotrauma 2017; 34:2518-2528. [DOI: 10.1089/neu.2016.4909] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|