1
|
Ding D, Yang M, Zheng X, Zhao M. Discovery of KDM5D as a novel biomarker for traumatic brain injury identified through bioinformatics analysis. Front Immunol 2025; 16:1538561. [PMID: 40196131 PMCID: PMC11973351 DOI: 10.3389/fimmu.2025.1538561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Background and aim Traumatic brain injury (TBI) poses a significant burden on the global economy due to its poor treatment and prognosis. Current TBI markers do not comprehensively reflect the disease status. Therefore, identifying more meaningful biomarkers is beneficial for improving the prognosis and clinical treatment of TBI patients. Methods The gene expression profile of TBI was obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were subjected to enrichment analysis, and key potential genes were identified through the protein-protein interaction network and cytoHubba modules. ROC curves were used to construct diagnostic models for hub genes. Immunofluorescence experiments were conducted to detect the expression of candidate biomarkers in TBI rat models. Finally, we investigated the expression of TBI biomarkers in normal human organs and pan-cancer tumor tissues, and evaluated their correlation with immune infiltration in different tumors. Results A total of 44 DEGs were identified across four brain regions of TBI patients. Enrichment analysis revealed that these genes were primarily involved in intracellular and cell signal transduction pathways. Furthermore, three hub genes- RPS4Y1, KDM5D and NLGN4Y-were identified through different module analysis. The ROC curve diagnostic model also confirmed that these genes also have high diagnostic value in serum. Subsequently, the presence of Kdm5d was detected in the brain tissue of TBI rats through immunofluorescence experiments. Compared to normal rats, Kdm5d expression increased in the cortical area of TBI rats, with no significant change in the hippocampus area, aligning with observations in TBI patients. Immune infiltration analysis demonstrated changes in immune cell subsets in HIP and PCx, revealing that plasma cells and CD8 T cells were lowly expressed in TBI (HIP) and while neutrophils was under-expressed in TBI (PCx). Pan-cancer analysis indicated that KDM5D was significantly up-regulated in 23 cancers, down-regulated in 3 cancers, and significantly associated with immune infiltration in 10 cancers. Conclusion Based on the results of bioinformatics analysis and animal experiments, KDM5D serves as a potential biomarker for the diagnosis and prognosis of TBI. Additionally, research on KDM5D may develop into new serum markers, providing new indicators for further clinical liquid biopsy and aiding in the prevention of both TBI and tumors to a certain extent.
Collapse
Affiliation(s)
- Dengfeng Ding
- Medical Innovation Research Department, Chinese PLA General Hospital, Beijing, China
| | - Mengzhe Yang
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Xinou Zheng
- Medical Innovation Research Department, Chinese PLA General Hospital, Beijing, China
| | - Ming Zhao
- Department of Neurosurgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Kadry MO, Ali H. Fischer's ratio and DNA damage in hypoxemia-induced brain injury in rat model: prophylactic role of quercetin and mexamine supplementation. PLoS One 2025; 20:e0319898. [PMID: 40100888 PMCID: PMC11918368 DOI: 10.1371/journal.pone.0319898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 02/03/2025] [Indexed: 03/20/2025] Open
Abstract
Hypoxemia brain injuries arise when the brain's oxygen supply is restricted. Brain cells gradually die and become impaired as a result of the restricted oxygen flow a diversity of signaling pathways are involved in the pathophysiology of brain damage. One of the main concerns when examining the rate of protein breakdown is the measurement of the serum amino acid ratio. Valine, leucine, and isoleucine make up branched-chain amino acids, while phenylalanine and tyrosine make up aromatic amino acids. A vital tool for assessing the severity of hypoxemia is Fischer's ratio. The goal of this article is to determine how quercetin (QUR) and/or mexamine (MEX) prevented synfat (SN)-induced brain damage in a rat models. It also aimed to elucidate the various cross-linked inflammatory pathways, DNA damage, and Fischer's ratio. Following QUR and MEX therapy, synfat-induced hypoxemia. Hemoglobin (Hb) levels were markedly reduced by synfat-intoxication, and oxidative stress and inflammatory biomarkers, including TNF-??, MDA, interleukin-6 (IL-6), and C -reactive protein (CRP), were elevated. Hemoglobin levels, oxidative stress biomarkers, and the aberrant expression of pro-inflammatory cytokines were all altered by QUR and/or MEX therapy. Similarly, the concentration of γ-aminobutyric acid, serotonine, noradrenaline, and intropin in cerebral tissue is restricted. Similarly, the COMET assay and 8-oxo-7,8-dihydro-2'-deoxyguanosine analysis (8-oxodG) demonstrated that QUR and MEX potentially altered synfat-induced brain DNA damage. The results confirmed the potential impact of this combined strategy as a powerful therapy for brain hypoxemia, concluding that treatment via QUR with MEX was superior therapy in modulating synfat-triggered cerebral injury.
Collapse
Affiliation(s)
- Mai O Kadry
- Therapeutic Chemistry Department, National Research Centre, El Buhouth St., Dokki, Egypt
| | - Hanaa Ali
- Department of Genetics and Cytology, National Research Centre, Dokki, Egypt
| |
Collapse
|
3
|
Zhang Y, Savvidou M, Liaudanskaya V, Singh P, Fu Y, Nasreen A, Coe M, Kelly M, Snapper D, Wagner C, Gill J, Symes A, Patra A, Kaplan DL, Beheshti A, Georgakoudi I. Synergistic label-free fluorescence imaging and miRNA studies reveal dynamic human neuron-glial metabolic interactions following injury. SCIENCE ADVANCES 2024; 10:eadp1980. [PMID: 39661671 PMCID: PMC11633737 DOI: 10.1126/sciadv.adp1980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Neuron-glial cell interactions following traumatic brain injury (TBI) determine the propagation of damage and long-term neurodegeneration. Spatiotemporally heterogeneous cytosolic and mitochondrial metabolic pathways are involved, leading to challenges in developing effective diagnostics and treatments. An engineered three-dimensional brain tissue model comprising human neurons, astrocytes, and microglia is used in combination with label-free, two-photon imaging and microRNA studies to characterize metabolic interactions between glial and neuronal cells over 72 hours following impact injury. We interpret multiparametric, quantitative, optical metabolic assessments in the context of microRNA gene set analysis and identify distinct metabolic changes in neurons and glial cells. Glycolysis, nicotinamide adenine dinucleotide phosphate (reduced form) and glutathione synthesis, fatty acid synthesis, and oxidation are mobilized within glial cells to mitigate the impacts of initial enhancements in oxidative phosphorylation and fatty acid oxidation within neurons, which lack robust antioxidant defenses. This platform enables enhanced understanding of mechanisms that may be targeted to improve TBI diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Maria Savvidou
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Volha Liaudanskaya
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Pramesh Singh
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA
| | - Yuhang Fu
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Amreen Nasreen
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Marly Coe
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Marilyn Kelly
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Dustin Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Chelsea Wagner
- School of Nursing, Johns Hopkins University, 525 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Jessica Gill
- School of Nursing, Johns Hopkins University, 525 N. Wolfe Street, Baltimore, MD 21205, USA
- Department of Neurology, School of Medicine, Johns Hopkins University, 525 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Aviva Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Abani Patra
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Afshin Beheshti
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Program in Cell, Molecular, and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Dartmouth Cancer Center, Dartmouth Hitchcock Medical Center, Lebanon NH 03766, USA
| |
Collapse
|
4
|
Park J, Lee SH, Shin D, Kim Y, Kim YS, Seong MY, Lee JJ, Seo HG, Cho WS, Ro YS, Kim Y, Oh BM. Multiplexed Quantitative Proteomics Reveals Proteomic Alterations in Two Rodent Traumatic Brain Injury Models. J Proteome Res 2024; 23:249-263. [PMID: 38064581 DOI: 10.1021/acs.jproteome.3c00544] [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] [Indexed: 01/06/2024]
Abstract
In many cases of traumatic brain injury (TBI), conspicuous abnormalities, such as scalp wounds and intracranial hemorrhages, abate over time. However, many unnoticeable symptoms, including cognitive, emotional, and behavioral dysfunction, often last from several weeks to years after trauma, even for mild injuries. Moreover, the cause of such persistence of symptoms has not been examined extensively. Recent studies have implicated the dysregulation of the molecular system in the injured brain, necessitating an in-depth analysis of the proteome and signaling pathways that mediate the consequences of TBI. Thus, in this study, the brain proteomes of two TBI models were examined by quantitative proteomics during the recovery period to determine the molecular mechanisms of TBI. Our results show that the proteomes in both TBI models undergo distinct changes. A bioinformatics analysis demonstrated robust activation and inhibition of signaling pathways and core proteins that mediate biological processes after brain injury. These findings can help determine the molecular mechanisms that underlie the persistent effects of TBI and identify novel targets for drug interventions.
Collapse
Affiliation(s)
- Junho Park
- Department of Pharmacology, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Research Institute for Basic Medical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Seung Hak Lee
- Department of Rehabilitation Medicine, Asan Medical Center, 88 Olympic-Ro 43-Gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Dongyoon Shin
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Yeongshin Kim
- Department of Life Science, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Young Sik Kim
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Min Yong Seong
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Jin Joo Lee
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Won-Sang Cho
- Department of Neurosurgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Young Sun Ro
- Department of Emergency Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Youngsoo Kim
- Proteomics Research Team, CHA Future Medicine Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Department of Life Science, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Institute of Aging, Seoul National University College of Medicine, 71 Ihwajang-gil, Jongno-gu, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- National Traffic Injury Rehabilitation Hospital, 260 Jungang-ro, Yangpyeong-gun 12564, Gyeonggi-do, Republic of Korea
| |
Collapse
|
5
|
Brazdzionis J, Radwan MM, Thankam F, Rajesh Lal M, Baron D, Connett DA, Agrawal DK, Miulli DE. A Swine Model of Traumatic Brain Injury: Effects of Neuronally Generated Electromagnetic Fields and Electromagnetic Field Stimulation on Traumatic Brain Injury-Related Changes. Cureus 2023; 15:e42544. [PMID: 37637613 PMCID: PMC10460141 DOI: 10.7759/cureus.42544] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Background and objective Traumatic brain injury (TBI) has been associated with aberrations in neural circuitry attributable to the pathology resulting in electromagnetic field (EMF) changes. These changes have been evaluated in a variety of settings including through novel induction sensors with an ultra-portable shielded helmet and EMF channels with differences identified by comparing pre-injury and post-injury states. Modulation of the EMF has undergone cursory evaluation in neurologic conditions but has not yet been fully evaluated for clinical effects in treatment. Target EMF stimulation using EMF-related changes preoperatively to postoperatively has not yet been attempted and has not been completed using induction sensor technology. Our objectives in this study were twofold: we wanted to test the hypothesis that targeted stimulation using an EMF signal generator and stimulator to abnormal thresholds identified by real-time measurement of EMFs may enable early resolution of EMF changes and treatment of the TBI as modeled through controlled cortical impact (CCI); we also aimed to assess the feasibility of attempting this using real-time measurements with an EMF shielded helmet with EMF channels and non-contact, non-invasive induction sensors with attached EMF transmitters in real-time. Methods A singular Yucatan miniswine was obtained and baseline EMF recordings were obtained. A CCI of TBI and postoperative assessment of cortical EMF in a non-invasive, non-contact fashion were completed. Alterations in EMF were evaluated and EMF stimulation using those abnormal frequencies was completed using multiple treatments involving three minutes of EMF stimulation at abnormal frequencies. Stimulation thresholds of 2.5 Hz, 3.5 Hz, and 5.5 Hz with 1 V signal intensity were evaluated using sinusoidal waves. Additionally, stimulation thresholds using differing offsets to the sine wave at -500 mV, 0 mV, and 500 mv were assessed. Daily EMF and post-stimulation EMF measurements were recorded. EMF patterns were then assessed using an artificial intelligence (AI) model. Results AI modeling appropriately identified differences in EMF signal in pre-injury, post-injury, and post-stimulation states. EMF stimulation using a positive offset of 500 mV appeared to have maximal beneficial effects in return to baseline. Similarly targeted stimulation using thresholds of 2.5 Hz and 5.5 Hz with a positive 500 mV offset at 1 V allowed for recovery of EMF patterns post-injury towards patterns seen in baseline EMF measurements on stimulation day seven (postoperative day 17). Conclusion Stimulation of neural circuits with targeted EMF in a sinusoidal pattern with targeted thresholds after measurement with induction sensors with shielding isolated to a Mu-metal and copper mesh helmet and EMF channels is efficacious in promoting neuronal circuit recovery to preoperative baselines in the TBI miniswine model. Similarly, our findings confirm the appropriateness of this translational model in the evaluation of brain neuronal circuit EMF and that preoperative and post-trauma differences can be appropriately assessed with this technology.
Collapse
Affiliation(s)
- James Brazdzionis
- Neurosurgery, Riverside University Health System Medical Center, Moreno Valley, USA
| | - Mohamed M Radwan
- Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Finosh Thankam
- Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Merlin Rajesh Lal
- Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - David Baron
- Psychiatry and Behavioral Sciences, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - David A Connett
- Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Devendra K Agrawal
- Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Dan E Miulli
- Neurosurgery, Riverside University Health System Medical Center, Moreno Valley, USA
| |
Collapse
|
6
|
Zhu Z, Huang X, Du M, Wu C, Fu J, Tan W, Wu B, Zhang J, Liao ZB. Recent advances in the role of miRNAs in post-traumatic stress disorder and traumatic brain injury. Mol Psychiatry 2023; 28:2630-2644. [PMID: 37340171 PMCID: PMC10615752 DOI: 10.1038/s41380-023-02126-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/12/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023]
Abstract
Post-traumatic stress disorder (PTSD) is usually considered a psychiatric disorder upon emotional trauma. However, with the rising number of conflicts and traffic accidents around the world, the incidence of PTSD has skyrocketed along with traumatic brain injury (TBI), a complex neuropathological disease due to external physical force and is also the most common concurrent disease of PTSD. Recently, the overlap between PTSD and TBI is increasingly attracting attention, as it has the potential to stimulate the emergence of novel treatments for both conditions. Of note, treatments exploiting the microRNAs (miRNAs), a well-known class of small non-coding RNAs (ncRNAs), have rapidly gained momentum in many nervous system disorders, given the miRNAs' multitudinous and key regulatory role in various biological processes, including neural development and normal functioning of the nervous system. Currently, a wealth of studies has elucidated the similarities of PTSD and TBI in pathophysiology and symptoms; however, there is a dearth of discussion with respect to miRNAs in both PTSD and TBI. In this review, we summarize the recent available studies of miRNAs in PTSD and TBI and discuss and highlight promising miRNAs therapeutics for both conditions in the future.
Collapse
Affiliation(s)
- Ziyu Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xuekang Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mengran Du
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chenrui Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiayuanyuan Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weilin Tan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Biying Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jie Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Z B Liao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
7
|
Brazdzionis J, Radwan MM, Thankam FG, Rajesh Lal M, Baron D, Connett DA, Agrawal DK, Miulli DE. A Swine Model of Changes in the Neuronal Electromagnetic Field After Traumatic Brain Injury: A Pilot Study. Cureus 2023; 15:e41763. [PMID: 37575822 PMCID: PMC10416555 DOI: 10.7759/cureus.41763] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Background Traumatic brain injury (TBI) is a global cause of disability and mortality. Treatment depends on mitigation of secondary injury resulting in axonal injury, necrosis, brain dysfunction, and disruption of electrical and chemical signaling in neural circuits. To better understand TBI, translational models are required to study physiology, diagnostics, and treatments in homologous species, such as swine. Electromagnetic fields (EMFs) from altered neural circuits can be measured and historically have been reliant on expensive shielding and supercooling in magnetoencephalography. Using proprietary induction sensors, it has been found that a non-invasive, non-contact approach with an engineered Mu-metal and copper mesh-shielded helmet effectively measures EMFs. This has not yet been investigated in swine models. We wished to evaluate the efficacy of this technology to assess TBI-dependent EMF changes in swine to describe the efficacy of these sensors and this model using a gravity-dependent controlled cortical impact (CCI). Methods A Yucatan miniswine was evaluated using non-contact, non-invasive proprietary induction sensors with an engineered dual-layer Mu-metal and interlaced copper mesh helmet with sensors within EMF channels connected to a helmet. Swine EMF recordings were obtained prior to induced gravity-dependent CCI followed by post-TBI measurements. Behavioral changes and changes in EMF measurements were assessed. EMF measurements were evaluated with an artificial intelligence (AI) model. Results Differences between room "noise" EMF measurements and pre-TBI swine electromagnetic field measurements were identified. Morphological characteristics between pre-injury and post-injury measurements were noted. AI modeling differentiated pre-injury and post-injury patterns in the swine EMF. Frequently identified frequencies seen post-injury were peaks at 2.5 Hz and 6.5 Hz and a valley at 11 Hz. The AI model identified less changes in the slope and thus decreased variation of EMF measurements post-TBI between 4.5 Hz and 7 Hz. Conclusions For the first time, it was identified that cortical function in a swine can be appropriately measured using novel induction sensors and shielding isolated to a helmet and EMF channels. The swine model can be appropriately differentiated from the external noise signal with identifiably different pre-injury and post-injury EMFs. Patterns can be recognized within the post-injury EMF due to altered neural circuits that can be measured using these sensors continuously, non-invasively, and in real time.
Collapse
Affiliation(s)
- James Brazdzionis
- Neurosurgery, Riverside University Health System Medical Center, Moreno Valley, USA
| | - Mohamed M Radwan
- Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Finosh G Thankam
- Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Merlin Rajesh Lal
- Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - David Baron
- Psychiatry and Behavioral Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - David A Connett
- Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Devendra K Agrawal
- Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, USA
| | - Dan E Miulli
- Neurosurgery, Riverside University Health System Medical Center, Moreno Valley, USA
| |
Collapse
|
8
|
Díaz MM, Tsenkina Y, Arizanovska D, Mehlen P, Liebl DJ. DCC/netrin-1 regulates cell death in oligodendrocytes after brain injury. Cell Death Differ 2023; 30:397-406. [PMID: 36456775 PMCID: PMC9950151 DOI: 10.1038/s41418-022-01091-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Hallmark pathological features of brain trauma are axonal degeneration and demyelination because myelin-producing oligodendrocytes (OLs) are particularly vulnerable to injury-induced death signals. To reveal mechanisms responsible for this OL loss, we examined a novel class of "death receptors" called dependence receptors (DepRs). DepRs initiate pro-death signals in the absence of their respective ligand(s), yet little is known about their role after injury. Here, we investigated whether the deleted in colorectal cancer (DCC) DepR contributes to OL loss after brain injury. We found that administration of its netrin-1 ligand is sufficient to block OL cell death. We also show that upon acute injury, DCC is upregulated while netrin-1 is downregulated in perilesional tissues. Moreover, after genetically silencing pro-death activity using DCCD1290N mutant mice, we observed greater OL survival, greater myelin integrity, and improved motor function. Our findings uncover a novel role for the netrin-1/DCC pathway in regulating OL loss in the traumatically injured brain.
Collapse
Affiliation(s)
- Madelen M Díaz
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Yanina Tsenkina
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dena Arizanovska
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory - Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Université de Lyon1, Lyon, France.
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
9
|
Logan A, Belli A, Di Pietro V, Tavazzi B, Lazzarino G, Mangione R, Lazzarino G, Morano I, Qureshi O, Bruce L, Barnes NM, Nagy Z. The mechanism of action of a novel neuroprotective low molecular weight dextran sulphate: New platform therapy for neurodegenerative diseases like Amyotrophic Lateral Sclerosis. Front Pharmacol 2022; 13:983853. [PMID: 36110516 PMCID: PMC9468270 DOI: 10.3389/fphar.2022.983853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/01/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Acute and chronic neurodegenerative diseases represent an immense socioeconomic burden that drives the need for new disease modifying drugs. Common pathogenic mechanisms in these diseases are evident, suggesting that a platform neuroprotective therapy may offer effective treatments. Here we present evidence for the mode of pharmacological action of a novel neuroprotective low molecular weight dextran sulphate drug called ILB®. The working hypothesis was that ILB® acts via the activation of heparin-binding growth factors (HBGF). Methods: Pre-clinical and clinical (healthy people and patients with ALS) in vitro and in vivo studies evaluated the mode of action of ILB®. In vitro binding studies, functional assays and gene expression analyses were followed by the assessment of the drug effects in an animal model of severe traumatic brain injury (sTBI) using gene expression studies followed by functional analysis. Clinical data, to assess the hypothesized mode of action, are also presented from early phase clinical trials. Results: ILB® lengthened APTT time, acted as a competitive inhibitor for HGF-Glypican-3 binding, effected pulse release of heparin-binding growth factors (HBGF) into the circulation and modulated growth factor signaling pathways. Gene expression analysis demonstrated substantial similarities in the functional dysregulation induced by sTBI and various human neurodegenerative conditions and supported a cascading effect of ILB® on growth factor activation, followed by gene expression changes with profound beneficial effect on molecular and cellular functions affected by these diseases. The transcriptional signature of ILB® relevant to cell survival, inflammation, glutamate signaling, metabolism and synaptogenesis, are consistent with the activation of neuroprotective growth factors as was the ability of ILB® to elevate circulating levels of HGF in animal models and humans. Conclusion: ILB® releases, redistributes and modulates the bioactivity of HBGF that target disease compromised nervous tissues to initiate a cascade of transcriptional, metabolic and immunological effects that control glutamate toxicity, normalize tissue bioenergetics, and resolve inflammation to improve tissue function. This unique mechanism of action mobilizes and modulates naturally occurring tissue repair mechanisms to restore cellular homeostasis and function. The identified pharmacological impact of ILB® supports the potential to treat various acute and chronic neurodegenerative disease, including sTBI and ALS.
Collapse
Affiliation(s)
- Ann Logan
- Department of Biomedical Sciences, University of Warwick, Coventry, United Kingdom
- Axolotl Consulting Ltd., Droitwich, United Kingdom
- *Correspondence: Ann Logan,
| | - Antonio Belli
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Valentina Di Pietro
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Barbara Tavazzi
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Giacomo Lazzarino
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Renata Mangione
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Rome, Rome, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Catania, Italy
| | | | | | | | - Nicholas M. Barnes
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Zsuzsanna Nagy
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
10
|
Hung SY, Chung HY, Luo ST, Chu YT, Chen YH, MacDonald IJ, Chien SY, Kotha P, Yang LY, Hwang LL, Dun NJ, Chuang DM, Chen YH. Electroacupuncture improves TBI dysfunction by targeting HDAC overexpression and BDNF-associated Akt/GSK-3β signaling. Front Cell Neurosci 2022; 16:880267. [PMID: 36016833 PMCID: PMC9396337 DOI: 10.3389/fncel.2022.880267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Acupuncture or electroacupuncture (EA) appears to be a potential treatment in acute clinical traumatic brain injury (TBI); however, it remains uncertain whether acupuncture affects post-TBI histone deacetylase (HDAC) expression or impacts other biochemical/neurobiological events. Materials and methods We used behavioral testing, Western blot, and immunohistochemistry analysis to evaluate the cellular and molecular effects of EA at LI4 and LI11 in both weight drop-impact acceleration (WD)- and controlled cortical impact (CCI)-induced TBI models. Results Both WD- and CCI-induced TBI caused behavioral dysfunction, increased cortical levels of HDAC1 and HDAC3 isoforms, activated microglia and astrocytes, and decreased cortical levels of BDNF as well as its downstream mediators phosphorylated-Akt and phosphorylated-GSK-3β. Application of EA reversed motor, sensorimotor, and learning/memory deficits. EA also restored overexpression of HDAC1 and HDAC3, and recovered downregulation of BDNF-associated signaling in the cortex of TBI mice. Conclusion The results strongly suggest that acupuncture has multiple benefits against TBI-associated adverse behavioral and biochemical effects and that the underlying mechanisms are likely mediated by targeting HDAC overexpression and aberrant BDNF-associated Akt/GSK-3 signaling.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Division of Colorectal Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hsin-Yi Chung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Sih-Ting Luo
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yu-Ting Chu
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Iona J. MacDonald
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Szu-Yu Chien
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Peddanna Kotha
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Laboratory for Neural Repair, China Medical University Hospital, Taichung, Taiwan
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nae J. Dun
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, United States
| | - De-Maw Chuang
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan
- *Correspondence: Yi-Hung Chen,
| |
Collapse
|
11
|
Nidhi, Singh G, Valecha R, Shukla G, Kaushik D, Rahman MA, Gautam RK, Madan K, Mittal V, Singla RK. Neurobehavioral and Biochemical Evidences in Support of Protective Effect of Marrubiin (Furan Labdane Diterpene) from Marrubium vulgare Linn. and Its Extracts after Traumatic Brain Injury in Experimental Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4457973. [PMID: 35656476 PMCID: PMC9155918 DOI: 10.1155/2022/4457973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 02/05/2023]
Abstract
Traumatic brain injuries due to sudden accidents cause major physical and mental health problems and are one of the main reasons behind the mortality and disability of patients. Research on alternate natural sources could be a boon for the rehabilitation of poor TBI patients. The literature indicates the Marrubium vulgare Linn. and its secondary metabolite marrubiin (furan labdane diterpene) possess various pharmacological properties such as vasorelaxant, calcium channel blocker, antioxidant, and antiedematogenic activities. Hence, in the present research, both marrubiin and hydroalcoholic extracts of the plant were evaluated for their neuroprotective effect after TBI. The neurological severity score and oxidative stress parameters are significantly altered by the test samples. Moreover, the neurotransmitter analysis indicated a significant change in GABA and glutamate. The histopathological study also supported the observed results. The improved neuroprotective potential of the extract could be attributed to the presence of a large number of secondary metabolites including marrubiin.
Collapse
Affiliation(s)
- Nidhi
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Rekha Valecha
- Department of Pharmacy, Delhi Skill and Entrepreneurship University, New Delhi 110065, India
| | - Govind Shukla
- University College of Ayurveda, Dr. S. R. Rajasthan Ayurveda University, Jodhpur 342304, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Mohammad Akhlaquer Rahman
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Rupesh K. Gautam
- Department of Pharmacology, MM School of Pharmacy, MM University, Sadopur-Ambala 134007, India
| | - Kumud Madan
- Lloyd Institute of Management and Technology (Pharm.), Greater Noida, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- iGlobal Research and Publishing Foundation, New Delhi 110059, India
| |
Collapse
|
12
|
Ma Y, Liu Y, Ruan X, Liu X, Zheng J, Teng H, Shao L, Yang C, Wang D, Xue Y. Gene Expression Signature of Traumatic Brain Injury. Front Genet 2021; 12:646436. [PMID: 33859672 PMCID: PMC8042258 DOI: 10.3389/fgene.2021.646436] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/12/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Traumatic brain injury (TBI) is a brain function change caused by external forces, which is one of the main causes of death and disability worldwide. The aim of this study was to identify early diagnostic markers and potential therapeutic targets for TBI. Methods: Differences between TBI and controls in GSE89866 and GSE104687 were analyzed. The two groups of differentially expressed genes (DEGs) were combined for coexpression analysis, and the modules of interest were performed using enrichment analysis. Hub genes were identified by calculating area under curve (AUC) values of module genes, PPI network analysis, and functional similarity. Finally, the difference in immune cell infiltration between TBI and control was calculated by ssGSEA. Results: A total of 4,817 DEGs were identified in GSE89866 and 1,329 DEGs in GSE104687. They were clustered into nine modules. The genes of modules 1, 4, and 7 had the most crosstalk and were identified as important modules. Enrichment analysis revealed that they were mainly associated with neurodevelopment and immune inflammation. In the PPI network constructed by genes with top 50 AUC values in module genes, we identified the top 10 genes with the greatest connectivity. Among them, down-regulated RPL27, RPS4X, RPL23A, RPS15A, and RPL7A had similar functions and were identified as hub genes. In addition, DC and Tem were significantly up-regulated and down-regulated between TBI and control, respectively. Conclusion: We found that hub genes may have a diagnostic role for TBI. Molecular dysregulation mechanisms of TBI are associated with neurological and immune inflammation. These results may provide new ideas for the diagnosis and treatment of TBI.
Collapse
Affiliation(s)
- Yawen Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China.,Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Hao Teng
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Lianqi Shao
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China.,Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, China
| |
Collapse
|
13
|
Hou Q, Chen H, Liu Q, Yan X. FGF10 Attenuates Experimental Traumatic Brain Injury through TLR4/MyD88/NF-κB Pathway. Cells Tissues Organs 2021; 209:248-256. [PMID: 33440393 DOI: 10.1159/000511381] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/27/2020] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can induce neuronal apoptosis and neuroinflammation, resulting in substantial neuronal damage and behavioral disorders. Fibroblast growth factors (FGFs) have been shown to be critical mediators in tissue repair. However, the role of FGF10 in experimental TBI remains unknown. In this study, mice with TBI were established via weight-loss model and validated by increase of modified neurological severity scores (mNSS) and brain water content. Secondly, FGF10 levels were elevated in mice after TBI, whereas intraventricular injection of Ad-FGF10 decreased mNSS score and brain water content, indicating the remittance of neurological deficit and cerebral edema in TBI mice. In addition, neuronal damage could also be ameliorated by stereotactic injection of Ad-FGF10. Overexpression of FGF10 increased protein expression of Bcl-2, while it decreased Bax and cleaved caspase-3/PARP, and improved neuronal apoptosis in TBI mice. In addition, Ad-FGF10 relieved neuroinflammation induced by TBI and significantly reduced the level of interleukin 1β/6, tumor necrosis factor α, and monocyte chemoattractant protein-1. Moreover, Ad-FGF10 injection decreased the protein expression level of Toll-like receptor 4 (TLR4), MyD88, and phosphorylation of NF-κB (p-NF-κB), suggesting the inactivation of the TLR4/MyD88/NF-κB pathway. In conclusion, overexpression of FGF10 could ameliorate neurological deficit, neuronal apoptosis, and neuroinflammation through inhibition of the TLR4/MyD88/NF-κB pathway, providing a potential therapeutic strategy for brain injury in the future.
Collapse
Affiliation(s)
- Qinhan Hou
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Hongmou Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China,
| | - Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Xianlei Yan
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| |
Collapse
|
14
|
Rana A, Singh S, Deshmukh R, Kumar A. Pharmacological potential of tocopherol and doxycycline against traumatic brain injury-induced cognitive/motor impairment in rats. Brain Inj 2020; 34:1039-1050. [PMID: 32493074 DOI: 10.1080/02699052.2020.1772508] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/29/2020] [Accepted: 05/06/2020] [Indexed: 01/20/2023]
Abstract
Primary Objective The primary objective of this study was to explore the pharmacological potential of tocopherol and doxycycline against traumatic brain injury-induced cognitive/motor impairment in rats. Research Design Weight drop model of traumatic brain injury. Methods and Procedures After TBI, the animals were treated with doxycycline (50 and 100 mg/kg; p.o), tocopherol (5 and 10 mg/kg; p.o) alone and in combination as doxycycline and tocopherol (50 and 10 mg/kg; p.o) from 1st day to 28th day. The behavioral parameters were performed on a weekly basis from 1st day to 28th day. On 29th day, animals were sacrificed and striatum and cortex were homogenized for the estimation of biochemical (LPO, nitrite, and GSH), neuroinflammatory (IL-6, IL-1β, and TNF-α), and neurotransmitters (dopamine, norepinephrine, serotonin, GABA, and glutamate) analysis. Main Outcomes and Results Induction of TBI had significantly reduced locomotor activity, recognition memory, increased neuroinflammatory markers, and imbalance neurotransmitter levels. The treatment with doxycycline and tocopherol alone and in combination significantly attenuated locomotor activity, memory recognition, reduced neuroinflammation, preserved oxidative balance, and restored the level of neurotransmitters. Conclusions The neuroprotective effect of doxycycline and tocopherol might be due to its anti-inflammatory and free radical scavenging mechanisms. Abbreviations TBI: Traumatic brain injury; Doxy: Doxycycline; Toco: Tocopherol; LPO: Lipid peroxidation; MDA: Malondialdehyde; TNF-α: Tumor necrosis factor-alpha; IL-1b: Interleukin-1 beta; GSH: Glutathione; GABA: gamma-Aminobutyric acid.
Collapse
Affiliation(s)
- Arti Rana
- Department of Neuropharmacology, ISF College of Pharmacy , Moga, Punjab, India
| | - Shamsher Singh
- Department of Neuropharmacology, ISF College of Pharmacy , Moga, Punjab, India
| | - Rahul Deshmukh
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU) , Bathinda, Punjab, India
| | - Anoop Kumar
- Department of Neuropharmacology, ISF College of Pharmacy , Moga, Punjab, India
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli , Lucknow, UP, India
| |
Collapse
|
15
|
Keerthana S, Kumar A. Potential risks and benefits of zinc oxide nanoparticles: a systematic review. Crit Rev Toxicol 2020; 50:47-71. [PMID: 32186437 DOI: 10.1080/10408444.2020.1726282] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Zinc oxide nanoparticles are well-known metal oxide nanoparticles having numbers of applications in the field of cosmetology, medicine, and chemistry. However, the number of reports has indicated its toxicity also such as hepatotoxicity, pulmonary toxicity, neurotoxicity, and immunotoxicity. Thus, in this article, we have analyzed the potential risks and benefits of zinc oxide nanoparticles. The data related to risks and benefits of zinc oxide nanoparticles have been extracted from PubMed (from January 2007 to August 2019). A total of 3,892 studies have been published during this period regarding zinc oxide nanoparticles. On the basis of inclusion and exclusion criteria, 277 studies have been included for the analysis of risks and benefits. Emerging reports have indicated both risks and benefits of zinc oxide nanoparticles in concentration- and time-dependent manner under in vitro and in vivo conditions through different mechanism of action. In conclusion, zinc oxide nanoparticles could play a beneficial role in the treatment of various diseases but safety of these particles at particular effective concentration should be thoroughly evaluated.
Collapse
Affiliation(s)
- S Keerthana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| | - A Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| |
Collapse
|
16
|
Niu F, Qian K, Qi H, Zhao Y, Jiang Y, Sun M. Antiapoptotic and Anti-Inflammatory Effects of CPCGI in Rats with Traumatic Brain Injury. Neuropsychiatr Dis Treat 2020; 16:2975-2987. [PMID: 33324059 PMCID: PMC7733055 DOI: 10.2147/ndt.s281530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/16/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Compound porcine cerebroside and ganglioside injection (CPCGI) has been used for the treatment of certain brain disorders. Apoptosis and inflammation were reported to be involved in the pathogenesis of traumatic brain injury (TBI). Therefore, this study primarily investigated the effects of CPCGI on mitochondrial apoptotic signaling and PARP/NF-κB inflammatory signaling in a rat model of controlled cortical impact (CCI). MATERIALS AND METHODS CPCGI (0.6 mL/kg) was administered intraperitoneally 30 min after the induction of CCI. Mitochondrial apoptotic signaling and PARP/NF-κB inflammatory signaling were evaluated 24 h after CCI, and apoptotic cell death, neutrophil infiltration, and astrocyte and microglial activation were determined by TUNEL and immunofluorescent staining 3 days after CCI. RESULTS 1) CPCGI markedly enhanced cytosolic and mitochondrial Bcl-xL levels, the mitochondrial Bcl-xL/Bax ratio, and mitochondrial cytochrome (cyt) c levels and reduced cytosolic cyt c levels, caspase-3 activity, and nuclear AIF levels in brain tissues after traumatic injury; however, CPCGI had no significant effects on cytosolic or mitochondrial Bax levels, the cytosolic Bcl-xL/Bax ratio, or mitochondrial AIF levels. Moreover, CPCGI markedly reduced the TUNEL staining score in the contusion region. 2) CPCGI markedly reduced cytosolic and nuclear PARP levels and nuclear NF-κB p65 levels in brain tissues after traumatic injury but had no significant effect on cytosolic NF-κB p65 levels. In addition, CPCGI markedly reduced caspase-1 activity and the levels of caspase-1, ICAM-1, TNF-α, and IL-1β in brain tissues after traumatic injury and decreased the immunoreactivities of neutrophils, GFAP and Iba-1 in the region of CCI-induced contusion. CONCLUSION These data suggest that CPCGI can reduce brain injury due to trauma by suppressing both mitochondrial apoptotic signaling and PARP/NF-κB inflammatory signaling.
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
| | - 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
|
17
|
Li Z, Han K, Zhang D, Chen J, Xu Z, Hou L. The role of long noncoding RNA in traumatic brain injury. Neuropsychiatr Dis Treat 2019; 15:1671-1677. [PMID: 31303755 PMCID: PMC6605043 DOI: 10.2147/ndt.s206624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI), a mainly lethal and highly debilitating condition, is increasing worldwide. However, the underlying mechanism has not been fully elucidated and effective therapy is needed. Long noncoding RNAs (lncRNAs), which form a major class of noncoding RNAs, have emerged as novel targets for regulating physiological functions and mediating numerous neurological diseases. Notably, gene expression profile analyses have demonstrated aberrant changes in lncRNA expression in the cerebral cortex and hippocampus of rats, mice and human after TBI. lncRNAs may be associated with multiple pathophysiological processes following TBI and might play a crucial role in complications of TBI, such as traumatic optic neuropathy due to the regulation of specific signaling pathways. Some lncRNAs have also been found to be therapeutic targets for motor and cognitive recovery after TBI. lncRNAs may be promising biomarkers for TBI diagnosis, treatment, and prognosis prediction. However, further research isneeded to clarify the underlying mechanisms and therapeutic effects of lncRNAs on TBI. We review the current progress of studies on lncRNAs in TBI to draw more attention to their roles in this debilitating condition.
Collapse
Affiliation(s)
- Zhenxing Li
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Kaiwei Han
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Danfeng Zhang
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Jigang Chen
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Zheng Xu
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| |
Collapse
|