1
|
Main KL, Vakhtin AA, Zhuo J, Marion D, Adamson MM, Ashford JW, Gullapalli R, Furst AJ. An iterative ROC procedure identifies white matter tracts diagnostic for traumatic brain injury: an exploratory analysis in U.S. Veterans. Brain Inj 2025:1-19. [PMID: 40257224 DOI: 10.1080/02699052.2025.2492746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE Understanding the pathophysiology of traumatic brain injury (TBI) is crucial for effectively managing care. Diffusion tensor imaging (DTI) is an MRI technology that evaluates TBI pathology in brain white matter. However, DTI analysis generates numerous measures. Choosing between them remains an obstacle to clinical translation. In this study, we leveraged an iterative receiver operating characteristic (ROC) analysis to examine white matter tracts in a group of 380 Veterans, consisting of TBI (n = 243) and non-TBI patients (n = 137). METHOD For each participant, we obtained a whole brain tractography and extracted DTI measures from 50 tracts. The ROC analyzed these variables and produced decision trees of tracts diagnostic for TBI. We expanded our findings by applying jackknife resampling. This procedure removed potential outliers and yielded tracts not observed in the initial ROCs. Finally, we used logistic regression to confirm the tracts predicted TBI status. RESULTS Our results indicate ROC can identify tracts diagnostic for TBI. We also found that groups of tracts are more predictive than any single one. CONCLUSIONS These analyses show that ROC is a useful tool for exploring large, multivariate datasets and may inform the design of clinical algorithms.
Collapse
Affiliation(s)
- Keith L Main
- Traumatic Brain Injury Center of Excellence, Defense Health Agency, Silver Spring, Maryland, USA
| | - Andrei A Vakhtin
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Traumatic Brain Injury Division, Albuquerque, New Mexico, USA
| | - Jiachen Zhuo
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Donald Marion
- Traumatic Brain Injury Center of Excellence, Defense Health Agency, Silver Spring, Maryland, USA
| | - Maheen M Adamson
- Women's Operational Military Exposure Network, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Rehabilitation Services, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - J Wesson Ashford
- War Related Illness and Injury Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Rao Gullapalli
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ansgar J Furst
- War Related Illness and Injury Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
- Polytrauma System of Care, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
2
|
Miner AE, Groh JR, Farris C, Hattiangadi S, Cui A, Brickman AM, Alshikho M, Rabinovici GD, Rosen HJ, Cobigo Y, Asken B, Nowinski CJ, Bureau S, Shahrokhi F, Tripodis Y, Ly M, Altaras C, Lenio S, Stern RA, Rosen G, Kelley H, Huber BR, Stein TD, Mez J, McKee AC, Alosco ML. Does white matter and vascular injury from repetitive head impacts lead to a novel pattern on T2 FLAIR MRI? A hypothesis proposal and call for research. Alzheimers Dement 2025; 21:e70085. [PMID: 40145364 PMCID: PMC11947747 DOI: 10.1002/alz.70085] [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: 09/12/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 03/28/2025]
Abstract
The goal of this paper is to introduce the hypothesis that white matter (WM) and vascular injury are long-term consequences of repetitive head impacts (RHI) that result in a novel T2 fluid attenuated inversion recovery (FLAIR) magnetic resonance imaging pattern. A non-systematic literature review of autopsy and FLAIR studies of RHI-exposed adults was first conducted as a foundation for our hypothesis. A case series of RHI-exposed participants is presented to illustrate the unique FLAIR WM hyperintensities (WMH) pattern. Current literature shows a direct link between RHI and later-life WM/vascular neuropathologies, and that FLAIR WMH are associated with RHI, independent of modifiable vascular risk factors. Initial observations suggest a distinctive pattern of WMH in RHI-exposed participants, termed RHI-associated WMH (RHI-WMH). RHI-WMH defining features are as follows: (1) small, punctate, non-confluent, (2) spherical, and (3) proximal to the gray matter. Our hypothesis serves as a call for research to empirically validate RHI-WMH and clarify their biological and clinical correlates. HIGHLIGHTS: Repetitive head impacts (RHI) have been associated with later-life white matter (WM) and vascular neuropathologies. T2 FLAIR MRI of RHI-exposed participants reveals a potentially unique WM hyperintensity (WMH) pattern that is termed RHI-associated WMH (RHI-WMH). RHI-WMH are characterized as (1) small, punctate, and non-confluent, (2) spherical, and (3) proximal to the gray matter at an area anatomically susceptible to impact injury, such as the depths of the cortical sulci.
Collapse
|
3
|
Zhu WJ, Liu J, Li WH, Zhao ZY, Huang C, Yang JY, Lee HC, Zhao YJ. Gap junction intercellular communications regulates activation of SARM1 and protects against axonal degeneration. Cell Death Dis 2025; 16:13. [PMID: 39809779 PMCID: PMC11733139 DOI: 10.1038/s41419-025-07342-4] [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: 04/15/2024] [Revised: 12/10/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Sterile alpha and Toll/interleukin-1 receptor motif containing 1 (SARM1), a nicotinamide adenine dinucleotide (NAD)-utilizing enzyme, mediates axon degeneration (AxD) in various neurodegenerative diseases. It is activated by nicotinamide mononucleotide (NMN) to produce a calcium messenger, cyclic ADP-ribose (cADPR). This activity is blocked by elevated NAD level. Here, we verified this metabolic regulation in somatic HEK-293T cells by overexpressing NMN-adenyltransferase to elevate cellular NAD, which resulted not only in inhibition of their own SARM1 from producing cADPR but, surprisingly, also in the 5-10 neighboring wildtype cells in mixed cultures via connexin (Cx)-43. Direct visualization of gap junction intercellular communication (GJIC) was achieved by incubating cells with a permeant probe, PC11, which is converted by SARM1 into PAD11, a fluorescent NAD analog capable of traversing GJs. Extending the findings to dorsal root ganglion neurons, we further showed that CZ-48, a permeant NMN analog, or axotomy, activated SARM1 and the produced PAD11 was transferred to contacting axons via GJIC. The gap junction involved was identified as Cx36 instead. This neuronal GJIC was demonstrated to be functional, enabling healthy neurons to protect adjacent axotomized axons from degeneration. Inhibition of GJIC in mice by AAV-PHP.eB-mediated knockdown of Cx36 in brain induced neuroinflammation, which in turn activated SARM1 and resulted in axon degeneration as well as behavioral deficits. Our results demonstrate a novel intercellular regulation mechanism of SARM1 and reveal a protective role of healthy tissue against AxD induced by injury or neuroinflammation.
Collapse
Affiliation(s)
- Wen Jie Zhu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Jun Liu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Wan Hua Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Zhi Ying Zhao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Chongquan Huang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Jian Yuan Yang
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Hon Cheung Lee
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yong Juan Zhao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China.
| |
Collapse
|
4
|
Liao Y, Li Y, Wang L, Zhang Y, Sang L, Wang Q, Li P, Xiong K, Qiu M, Zhang J. The Injury Progression in Acute Blast-Induced Mild Traumatic Brain Injury in Rats Reflected by Diffusion Tensor Imaging and Immunohistochemical Examination. J Neurotrauma 2024; 41:2478-2492. [PMID: 38877821 DOI: 10.1089/neu.2023.0435] [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: 06/16/2024] Open
Abstract
Diffusion tensor imaging (DTI) has emerged as a promising neuroimaging tool for detecting blast-induced mild traumatic brain injury (bmTBI). However, lack of refined acute-phase monitoring and reliable imaging biomarkers hindered its clinical application in early diagnosis of bmTBI, leading to potential long-term disability of patients. In this study, we used DTI in a rat model of bmTBI generated by exposing to single lateral blast waves (151.16 and 349.75 kPa, lasting 47.48 ms) released in a confined bioshock tube, to investigate whole-brain DTI changes at 1, 3, and 7 days after injury. Combined assessment of immunohistochemical analysis, transmission electron microscopy, and behavioral readouts allowed for linking DTI changes to synchronous cellular damages and identifying stable imaging biomarkers. The corpus callosum (CC) and brainstem were identified as predominantly affected regions, in which reduced fractional anisotropy (FA) was detected as early as the first day after injury, with a maximum decline occurring at 3 days post-injury before returning to near normal levels by 7 days. Axial diffusivity (AD) values within the CC and brainstem also significantly reduced at 3 days post-injury. In contrast, the radial diffusivity (RD) in the CC showed acute elevation, peaking at 3 days after injury before normalizing by the 7-day time point. Damages to nerve fibers, including demyelination and axonal degeneration, progressed in lines with changes in DTI parameters, supporting a real-time macroscopic reflection of microscopic neuronal fiber injury by DTI. The most sensitive biomarker was identified as a decrease in FA, AD, and an increase in RD within the CC on the third day after injury, supporting the diagnostic utility of DTI in cases of bmTBI in the acute phase.
Collapse
Affiliation(s)
- Yalan Liao
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Yang Li
- Department of Medical Imaging, Air Force Hospital of Western Theater Command, Chengdu, China
| | - Li Wang
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Ye Zhang
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Linqiong Sang
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Qiannan Wang
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Pengyue Li
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Kunlin Xiong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingguo Qiu
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Jingna Zhang
- Department of Medical Imaging, College of Biomedical Engineering, Army Medical University, Chongqing, China
| |
Collapse
|
5
|
Gul E, Atalar MH, Atik I. Evaluation of the contralateral hemisphere with DWI in pediatric patients with Dyke-Davidoff-Masson syndrome. Acta Neurol Belg 2024; 124:911-918. [PMID: 38361171 DOI: 10.1007/s13760-024-02473-5] [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: 08/19/2023] [Accepted: 01/03/2024] [Indexed: 02/17/2024]
Abstract
INTRODUCTION Dyke-Davidoff-Masson Syndrome (DDMS) is a clinical syndrome that causes different clinical symptoms and is defined by volume decrement in one cerebral hemisphere. In this study, we aimed to evaluate the involvement of the normal-appearing contralateral hemisphere in 16 pediatric patients with DDMS using diffusion-weighted imaging (DWI). MATERIALS AND METHODS Brain MRIs were retrospectively reviewed between January 2014 and January 2023. Sixteen pediatric patients radiologically compatible with DDMS were included in the study. Sixteen children who had undergone brain MRI, most commonly for headaches and whose MRI findings had been completely normal, were included as the control group. Apparent diffusion coefficient (ADC) values of the deep gray and white matter of the normal-appearing hemisphere in the patient group were calculated and compared with that of the control group. RESULTS The ADC values of the gray and white matters of the patient and control groups were not statistically different. However, in the patient group, the ADC values of the gray and white matters in males were remarkably lower than in females (p = 0.038, p = 0.037, respectively). CONCLUSION The difference in the ADC values of the contralateral hemisphere between females and males in the patient group suggests that the normal-appearing hemisphere may have been affected by DDMS. Although, the exact mechanism of this effect is not known. Therefore, in patients with DDMS, contralateral hemisphere involvement in cerebral hemiatrophy and hemispherectomy should be evaluated clinically and radiologically.
Collapse
Affiliation(s)
- Enes Gul
- Sivas Cumhuriyet Universitesi, Sivas, Sivas, Turkey.
| | | | - Irfan Atik
- Sivas Cumhuriyet Universitesi, Sivas, Sivas, Turkey
| |
Collapse
|
6
|
Zhao Y, Zhou YG, Chen JF. Targeting the adenosine A 2A receptor for neuroprotection and cognitive improvement in traumatic brain injury and Parkinson's disease. Chin J Traumatol 2024; 27:125-133. [PMID: 37679245 PMCID: PMC11138351 DOI: 10.1016/j.cjtee.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/25/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Adenosine exerts its dual functions of homeostasis and neuromodulation in the brain by acting at mainly 2 G-protein coupled receptors, called A1 and A2A receptors. The adenosine A2A receptor (A2AR) antagonists have been clinically pursued for the last 2 decades, leading to final approval of the istradefylline, an A2AR antagonist, for the treatment of OFF-Parkinson's disease (PD) patients. The approval paves the way to develop novel therapeutic methods for A2AR antagonists to address 2 major unmet medical needs in PD and traumatic brain injury (TBI), namely neuroprotection or improving cognition. In this review, we first consider the evidence for aberrantly increased adenosine signaling in PD and TBI and the sufficiency of the increased A2AR signaling to trigger neurotoxicity and cognitive impairment. We further discuss the increasing preclinical data on the reversal of cognitive deficits in PD and TBI by A2AR antagonists through control of degenerative proteins and synaptotoxicity, and on protection against TBI and PD pathologies by A2AR antagonists through control of neuroinflammation. Moreover, we provide the supporting evidence from multiple human prospective epidemiological studies which revealed an inverse relation between the consumption of caffeine and the risk of developing PD and cognitive decline in aging population and Alzheimer's disease patients. Collectively, the convergence of clinical, epidemiological and experimental evidence supports the validity of A2AR as a new therapeutic target and facilitates the design of A2AR antagonists in clinical trials for disease-modifying and cognitive benefit in PD and TBI patients.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yuan-Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325035, Zhejiang Province, China.
| |
Collapse
|
7
|
Green TRF, Carey SD, Mannino G, Craig JA, Rowe RK, Zielinski MR. Sleep, inflammation, and hemodynamics in rodent models of traumatic brain injury. Front Neurosci 2024; 18:1361014. [PMID: 38426017 PMCID: PMC10903352 DOI: 10.3389/fnins.2024.1361014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Traumatic brain injury (TBI) can induce dysregulation of sleep. Sleep disturbances include hypersomnia and hyposomnia, sleep fragmentation, difficulty falling asleep, and altered electroencephalograms. TBI results in inflammation and altered hemodynamics, such as changes in blood brain barrier permeability and cerebral blood flow. Both inflammation and altered hemodynamics, which are known sleep regulators, contribute to sleep impairments post-TBI. TBIs are heterogenous in cause and biomechanics, which leads to different molecular and symptomatic outcomes. Animal models of TBI have been developed to model the heterogeneity of TBIs observed in the clinic. This review discusses the intricate relationship between sleep, inflammation, and hemodynamics in pre-clinical rodent models of TBI.
Collapse
Affiliation(s)
- Tabitha R. F. Green
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Sean D. Carey
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA, United States
| | - Grant Mannino
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - John A. Craig
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
| | - Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Mark R. Zielinski
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA, United States
| |
Collapse
|
8
|
Alexandris AS, Koliatsos VE. NAD +, Axonal Maintenance, and Neurological Disease. Antioxid Redox Signal 2023; 39:1167-1184. [PMID: 37503611 PMCID: PMC10715442 DOI: 10.1089/ars.2023.0350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 07/29/2023]
Abstract
Significance: The remarkable geometry of the axon exposes it to unique challenges for survival and maintenance. Axonal degeneration is a feature of peripheral neuropathies, glaucoma, and traumatic brain injury, and an early event in neurodegenerative diseases. Since the discovery of Wallerian degeneration (WD), a molecular program that hijacks nicotinamide adenine dinucleotide (NAD+) metabolism for axonal self-destruction, the complex roles of NAD+ in axonal viability and disease have become research priority. Recent Advances: The discoveries of the protective Wallerian degeneration slow (WldS) and of sterile alpha and TIR motif containing 1 (SARM1) activation as the main instructive signal for WD have shed new light on the regulatory role of NAD+ in axonal degeneration in a growing number of neurological diseases. SARM1 has been characterized as a NAD+ hydrolase and sensor of NAD+ metabolism. The discovery of regulators of nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) proteostasis in axons, the allosteric regulation of SARM1 by NAD+ and NMN, and the existence of clinically relevant windows of action of these signals has opened new opportunities for therapeutic interventions, including SARM1 inhibitors and modulators of NAD+ metabolism. Critical Issues: Events upstream and downstream of SARM1 remain unclear. Furthermore, manipulating NAD+ metabolism, an overdetermined process crucial in cell survival, for preventing the degeneration of the injured axon may be difficult and potentially toxic. Future Directions: There is a need for clarification of the distinct roles of NAD+ metabolism in axonal maintenance as contrasted to WD. There is also a need to better understand the role of NAD+ metabolism in axonal endangerment in neuropathies, diseases of the white matter, and the early stages of neurodegenerative diseases of the central nervous system. Antioxid. Redox Signal. 39, 1167-1184.
Collapse
Affiliation(s)
| | - Vassilis E. Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Lu Q, Zhang Y, Botchway BOA, Huang M, Liu X. Syntaphilin Inactivation Can Enhance Axonal Mitochondrial Transport to Improve Spinal Cord Injury. Mol Neurobiol 2023; 60:6556-6565. [PMID: 37458986 DOI: 10.1007/s12035-023-03494-6] [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: 02/25/2023] [Accepted: 07/08/2023] [Indexed: 09/28/2023]
Abstract
Mitochondria are important organelle of eukaryotic cells. They consists of a large number of different proteins that provide most of the ATP and supply power for the growth, function, and regeneration of neurons. Therefore, smitochondrial transport ensures that adequate ATP is supplied for metabolic activities. Spinal cord injury (SCI), a detrimental condition, has high morbidity and mortality rates. Currently, the available treatments only provide symptomatic relief for long-term disabilities. Studies have implicated mitochondrial transport as a critical factor in axonal regeneration. Hence, enhancing mitochondrial transports could be beneficial for ameliorating SCI. Syntaphilin (Snph) is a mitochondrial docking protein that acts as a "static anchor," and its inhibition enhances mitochondrial transports. Therefore, Snph as a key mediator of mitochondrial transports, may contribute to improving axonal regeneration following SCI. Herein, we examine Snph's biological effects and its relation to mitochondrial pathway. Then, we elaborate on mitochondrial transports after SCI, the possible role of Snph in SCI, and some possible therapeutic approaches by Snph.
Collapse
Affiliation(s)
- Qicheng Lu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
- Bupa Cromwell Hospital, London, UK
| | - Min Huang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
10
|
Brennan DJ, Duda J, Ware JB, Whyte J, Choi JY, Gugger J, Focht K, Walter AE, Bushnik T, Gee JC, Diaz‐Arrastia R, Kim JJ. Spatiotemporal profile of atrophy in the first year following moderate-severe traumatic brain injury. Hum Brain Mapp 2023; 44:4692-4709. [PMID: 37399336 PMCID: PMC10400790 DOI: 10.1002/hbm.26410] [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: 12/12/2022] [Revised: 06/04/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023] Open
Abstract
Traumatic brain injury (TBI) triggers progressive neurodegeneration resulting in brain atrophy that continues months-to-years following injury. However, a comprehensive characterization of the spatial and temporal evolution of TBI-related brain atrophy remains incomplete. Utilizing a sensitive and unbiased morphometry analysis pipeline optimized for detecting longitudinal changes, we analyzed a sample consisting of 37 individuals with moderate-severe TBI who had primarily high-velocity and high-impact injury mechanisms. They were scanned up to three times during the first year after injury (3 months, 6 months, and 12 months post-injury) and compared with 33 demographically matched controls who were scanned once. Individuals with TBI already showed cortical thinning in frontal and temporal regions and reduced volume in the bilateral thalami at 3 months post-injury. Longitudinally, only a subset of cortical regions in the parietal and occipital lobes showed continued atrophy from 3 to 12 months post-injury. Additionally, cortical white matter volume and nearly all deep gray matter structures exhibited progressive atrophy over this period. Finally, we found that disproportionate atrophy of cortex along sulci relative to gyri, an emerging morphometric marker of chronic TBI, was present as early as 3 month post-injury. In parallel, neurocognitive functioning largely recovered during this period despite this pervasive atrophy. Our findings demonstrate msTBI results in characteristic progressive neurodegeneration patterns that are divergent across regions and scale with the severity of injury. Future clinical research using atrophy during the first year of TBI as a biomarker of neurodegeneration should consider the spatiotemporal profile of atrophy described in this study.
Collapse
Affiliation(s)
- Daniel J. Brennan
- CUNY Neuroscience Collaborative, The Graduate CenterCity University of New YorkNew YorkNew YorkUnited States
- Department of Molecular, Cellular, and Biomedical SciencesCUNY School of Medicine, The City College of New YorkNew YorkNew YorkUnited States
| | - Jeffrey Duda
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
- Penn Image Computing and Science LaboratoryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUnited States
| | - Jeffrey B. Ware
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - John Whyte
- Moss Rehabilitation Research Institute, Einstein Healthcare NetworkElkins ParkPennsylvaniaUnited States
| | - Joon Yul Choi
- Department of Molecular, Cellular, and Biomedical SciencesCUNY School of Medicine, The City College of New YorkNew YorkNew YorkUnited States
- Department of Biomedical EngineeringYonsei UniversityWonjuRepublic of Korea
| | - James Gugger
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - Kristen Focht
- Widener University School for Graduate Clinical PsychologyChesterPennsylvaniaUnited States
| | - Alexa E. Walter
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - Tamara Bushnik
- NYU Grossman School of MedicineNew YorkNew YorkUnited States
| | - James C. Gee
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
- Penn Image Computing and Science LaboratoryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUnited States
| | - Ramon Diaz‐Arrastia
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - Junghoon J. Kim
- CUNY Neuroscience Collaborative, The Graduate CenterCity University of New YorkNew YorkNew YorkUnited States
- Department of Molecular, Cellular, and Biomedical SciencesCUNY School of Medicine, The City College of New YorkNew YorkNew YorkUnited States
| |
Collapse
|
11
|
Clemente A, Attyé A, Renard F, Calamante F, Burmester A, Imms P, Deutscher E, Akhlaghi H, Beech P, Wilson PH, Poudel G, Domínguez D JF, Caeyenberghs K. Individualised profiling of white matter organisation in moderate-to-severe traumatic brain injury patients. Brain Res 2023; 1806:148289. [PMID: 36813064 DOI: 10.1016/j.brainres.2023.148289] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/22/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND AND PURPOSE Approximately 65% of moderate-to-severe traumatic brain injury (m-sTBI) patients present with poor long-term behavioural outcomes, which can significantly impair activities of daily living. Numerous diffusion-weighted MRI studies have linked these poor outcomes to decreased white matter integrity of several commissural tracts, association fibres and projection fibres in the brain. However, most studies have focused on group-based analyses, which are unable to deal with the substantial between-patient heterogeneity in m-sTBI. As a result, there is increasing interest and need in conducting individualised neuroimaging analyses. MATERIALS AND METHODS Here, we generated a detailed subject-specific characterisation of microstructural organisation of white matter tracts in 5 chronic patients with m-sTBI (29 - 49y, 2 females), presented as a proof-of-concept. We developed an imaging analysis framework using fixel-based analysis and TractLearn to determine whether the values of fibre density of white matter tracts at the individual patient level deviate from the healthy control group (n = 12, 8F, Mage = 35.7y, age range 25 - 64y). RESULTS Our individualised analysis revealed unique white matter profiles, confirming the heterogenous nature of m-sTBI and the need of individualised profiles to properly characterise the extent of injury. Future studies incorporating clinical data, as well as utilising larger reference samples and examining the test-retest reliability of the fixel-wise metrics are warranted. CONCLUSIONS Individualised profiles may assist clinicians in tracking recovery and planning personalised training programs for chronic m-sTBI patients, which is necessary to achieve optimal behavioural outcomes and improved quality of life.
Collapse
Affiliation(s)
- Adam Clemente
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural, Health and Human Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, Australia.
| | - Arnaud Attyé
- CNRS LPNC UMR 5105, University of Grenoble Alpes, Grenoble, France; School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Félix Renard
- CNRS LPNC UMR 5105, University of Grenoble Alpes, Grenoble, France
| | - Fernando Calamante
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia; Sydney Imaging - The University of Sydney, Sydney, Australia
| | - Alex Burmester
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia
| | - Phoebe Imms
- Leonard Davis School of Gerontology, University of Southern California, Australia
| | - Evelyn Deutscher
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia
| | - Hamed Akhlaghi
- Emergency Department, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia; Department of Psychology, Faculty of Health, Deakin University, Australia
| | - Paul Beech
- Department of Radiology and Nuclear Medicine, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Peter H Wilson
- Development and Disability over the Lifespan Program, Healthy Brain and Mind Research Centre, School of Behavioural, Health and Human Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, Australia
| | - Govinda Poudel
- Mary MacKillop Institute for Health Research, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, Australia
| | - Juan F Domínguez D
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia
| | - Karen Caeyenberghs
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
12
|
Sabetta Z, Krishna G, Curry T, Adelson PD, Thomas TC. Aging with TBI vs. Aging: 6-month temporal profiles for neuropathology and astrocyte activation converge in behaviorally relevant thalamocortical circuitry of male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527058. [PMID: 36798182 PMCID: PMC9934568 DOI: 10.1101/2023.02.06.527058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Traumatic brain injury (TBI) manifests late-onset and persisting clinical symptoms with implications for sex differences and increased risk for the development of age-related neurodegenerative diseases. Few studies have evaluated chronic temporal profiles of neuronal and glial pathology that include sex as a biological variable. After experimental diffuse TBI, late-onset and persisting somatosensory hypersensitivity to whisker stimulation develops at one-month post-injury and persists to at least two months post-injury in male rats, providing an in vivo model to evaluate the temporal profile of pathology responsible for morbidity. Whisker somatosensation is dependent on signaling through the thalamocortical relays of the whisker barrel circuit made up of glutamatergic projections between the ventral posteromedial nucleus of the thalamus (VPM) and primary somatosensory barrel cortex (S1BF) with inhibitory (GABA) innervation from the thalamic reticular nucleus (TRN) to the VPM. To evaluate the temporal profiles of pathology, male and female Sprague Dawley rats ( n = 5-6/group) were subjected to sham surgery or midline fluid percussion injury (FPI). At 7-, 56-, and 168-days post-injury (DPI), brains were processed for amino-cupric silver stain and glial fibrillary acidic protein (GFAP) immunoreactivity, where pixel density of staining was quantified to determine the temporal profile of neuropathology and astrocyte activation in the VPM, S1BF, and TRN. FPI induced significant neuropathology in all brain regions at 7 DPI. At 168 DPI, neuropathology remained significantly elevated in the VPM and TRN, but returned to sham levels in the S1BF. GFAP immunoreactivity was increased as a function of FPI and DPI, with an FPI × DPI interaction in all regions and an FPI × Sex interaction in the S1BF. The interactions were driven by increased GFAP immunoreactivity in shams over time in the VPM and TRN. In the S1BF, GFAP immunoreactivity increased at 7 DPI and declined to age-matched sham levels by 168 DPI, while GFAP immunoreactivity in shams significantly increased between 7 and 168 days. The FPI × Sex interaction was driven by an overall greater level of GFAP immunoreactivity in FPI males compared to FPI females. Increased GFAP immunoreactivity was associated with an increased number of GFAP-positive soma, predominantly at 7 DPI. Overall, these findings indicate that FPI, time post-injury, sex, region, and aging with injury differentially contribute to chronic changes in neuronal pathology and astrocyte activation after diffuse brain injury. Thus, our results highlight distinct patterns of pathological alterations associated with the development and persistence of morbidity that supports chronic neuropathology, especially within the thalamus. Further, data indicate a convergence between TBI-induced and age-related pathology where further investigation may reveal a role for divergent astrocytic phenotypes associated with increased risk for neurodegenerative diseases.
Collapse
|
13
|
Mohammed FS, Omay SB, Sheth KN, Zhou J. Nanoparticle-based drug delivery for the treatment of traumatic brain injury. Expert Opin Drug Deliv 2023; 20:55-73. [PMID: 36420918 PMCID: PMC9983310 DOI: 10.1080/17425247.2023.2152001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Traumatic brain injuries (TBIs) impact the breadth of society and remain without any approved pharmacological treatments. Despite successful Phase II clinical trials, the failure of many Phase III clinical trials may be explained by insufficient drug targeting and retention, preventing the proper attainment of an observable dosage threshold. To address this challenge, nanoparticles can be functionalized to protect pharmacological payloads, improve targeted drug delivery to sites of injury, and can be combined with supportive scaffolding to improve secondary outcomes. AREAS COVERED This review briefly covers the pathophysiology of TBIs and their subtypes, the current pre-clinical and clinical management strategies, explores the common models of focal, diffuse, and mixed traumatic brain injury employed in experimental animals, and surveys the existing literature on nanoparticles developed to treat TBIs. EXPERT OPINION Nanoparticles are well suited to improve secondary outcomes as their multifunctionality and customizability enhance their potential for efficient targeted delivery, payload protection, increased brain penetration, low off-target toxicity, and biocompatibility in both acute and chronic timescales.
Collapse
Affiliation(s)
- Farrah S. Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Sacit Bulent Omay
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
- Department of Neurology, Yale University, New Haven, Connecticut, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Traumatic axonopathy in spinal tracts after impact acceleration head injury: Ultrastructural observations and evidence of SARM1-dependent axonal degeneration. Exp Neurol 2023; 359:114252. [PMID: 36244414 DOI: 10.1016/j.expneurol.2022.114252] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022]
Abstract
Traumatic axonal injury (TAI) and the associated axonopathy are common consequences of traumatic brain injury (TBI) and contribute to significant neurological morbidity. It has been previously suggested that TAI activates a highly conserved program of axonal self-destruction known as Wallerian degeneration (WD). In the present study, we utilize our well-established impact acceleration model of TBI (IA-TBI) to characterize the pathology of injured myelinated axons in the white matter tracks traversing the ventral, lateral, and dorsal spinal columns in the mouse and assess the effect of Sterile Alpha and TIR Motif Containing 1 (Sarm1) gene knockout on acute and subacute axonal degeneration and myelin pathology. In silver-stained preparations, we found that IA-TBI results in white matter pathology as well as terminal field degeneration across the rostrocaudal axis of the spinal cord. At the ultrastructural level, we found that traumatic axonopathy is associated with diverse types of axonal and myelin pathology, ranging from focal axoskeletal perturbations and focal disruption of the myelin sheath to axonal fragmentation. Several morphological features such as neurofilament compaction, accumulation of organelles and inclusions, axoskeletal flocculation, myelin degeneration and formation of ovoids are similar to profiles encountered in classical examples of WD. Other profiles such as excess myelin figures and inner tongue evaginations are more typical of chronic neuropathies. Stereological analysis of pathological axonal and myelin profiles in the ventral, lateral, and dorsal columns of the lower cervical cord (C6) segments from wild type and Sarm1 KO mice at 3 and 7 days post IA-TBI (n = 32) revealed an up to 90% reduction in the density of pathological profiles in Sarm1 KO mice after IA-TBI. Protection was evident across all white matter tracts assessed, but showed some variability. Finally, Sarm1 deletion ameliorated the activation of microglia associated with TAI. Our findings demonstrate the presence of severe traumatic axonopathy in multiple ascending and descending long tracts after IA-TBI with features consistent with some chronic axonopathies and models of WD and the across-tract protective effect of Sarm1 deletion.
Collapse
|
15
|
Sarkar A, Kumari N, Mukherjee P. The curious case of SARM1: Dr. Jekyll and Mr. Hyde in cell death and immunity? FEBS J 2023; 290:340-358. [PMID: 34710262 DOI: 10.1111/febs.16256] [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: 07/26/2021] [Revised: 09/21/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Sterile alpha and toll/interleukin-1 receptor motif-containing protein 1 (SARM1) was first identified as a novel ortholog of Drosophila protein CG7915 and was subsequently placed as the fifth member of the human TIR-containing adaptor protein. SARM1 holds a unique position in this family where, unlike other members, it downregulates NFκB activity in response to immunogenic stimulation, interacts with another member of the family, TRIF, to negatively regulate its function, and it also mediates cell death responses. Over the past decade, SARM1 has emerged as one of the primary mediators of programmed axonal degeneration and this robust regulation of axonal degeneration-especially in models of peripheral neuropathy and traumatic injury-makes it an attractive target for therapeutic intervention. The TIR domain of SARM1 possesses an intrinsic NADase activity resulting in cellular energy deficits within the axons, a striking deviation from its other family members of human TLR adaptors. Interestingly, the TIR NADase activity, as seen in SARM1, is also observed in several prokaryotic TIR-containing proteins where they are involved in immune evasion once within the host. Although the immune function of SARM1 is yet to be conclusively discerned, this closeness in function with the prokaryotic TIR-domain containing proteins, places it at an interesting juncture of evolution raising questions about its origin and function in cell death and immunity. In this review, we discuss how a conserved immune adaptor protein like SARM1 switches to a pro-neurodegenerative function and the evolutionarily significance of the process.
Collapse
Affiliation(s)
- Ankita Sarkar
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Nripa Kumari
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Piyali Mukherjee
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
16
|
Bratkowski M, Burdett TC, Danao J, Wang X, Mathur P, Gu W, Beckstead JA, Talreja S, Yang YS, Danko G, Park JH, Walton M, Brown SP, Tegley CM, Joseph PRB, Reynolds CH, Sambashivan S. Uncompetitive, adduct-forming SARM1 inhibitors are neuroprotective in preclinical models of nerve injury and disease. Neuron 2022; 110:3711-3726.e16. [PMID: 36087583 DOI: 10.1016/j.neuron.2022.08.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/06/2022] [Accepted: 08/10/2022] [Indexed: 12/15/2022]
Abstract
Axon degeneration is an early pathological event in many neurological diseases. The identification of the nicotinamide adenine dinucleotide (NAD) hydrolase SARM1 as a central metabolic sensor and axon executioner presents an exciting opportunity to develop novel neuroprotective therapies that can prevent or halt the degenerative process, yet limited progress has been made on advancing efficacious inhibitors. We describe a class of NAD-dependent active-site SARM1 inhibitors that function by intercepting NAD hydrolysis and undergoing covalent conjugation with the reaction product adenosine diphosphate ribose (ADPR). The resulting small-molecule ADPR adducts are highly potent and confer compelling neuroprotection in preclinical models of neurological injury and disease, validating this mode of inhibition as a viable therapeutic strategy. Additionally, we show that the most potent inhibitor of CD38, a related NAD hydrolase, also functions by the same mechanism, further underscoring the broader applicability of this mechanism in developing therapies against this class of enzymes.
Collapse
Affiliation(s)
| | - Thomas C Burdett
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Jean Danao
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Xidao Wang
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Prakhyat Mathur
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Weijing Gu
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | | | - Santosh Talreja
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Yu-San Yang
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Gregory Danko
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Jae Hong Park
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Mary Walton
- Chemistry Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Sean P Brown
- Chemistry Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | | | - Prem Raj B Joseph
- WuXi AppTec, Research Services Division, 6 Cedarbrook Drive, Cranbury, NJ 08512, USA
| | | | | |
Collapse
|
17
|
Brooks LJ, Simpson Ragdale H, Hill CS, Clements M, Parrinello S. Injury programs shape glioblastoma. Trends Neurosci 2022; 45:865-876. [PMID: 36089406 DOI: 10.1016/j.tins.2022.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/30/2022] [Accepted: 08/09/2022] [Indexed: 11/20/2022]
Abstract
Glioblastoma is the most common and aggressive primary brain cancer in adults and is almost universally fatal due to its stark therapeutic resistance. During the past decade, although survival has not substantially improved, major advances have been made in our understanding of the underlying biology. It has become clear that these devastating tumors recapitulate features of neurodevelopmental hierarchies which are influenced by the microenvironment. Emerging evidence also highlights a prominent role for injury responses in steering cellular phenotypes and contributing to tumor heterogeneity. This review highlights how the interplay between injury and neurodevelopmental programs impacts on tumor growth, invasion, and treatment resistance, and discusses potential therapeutic considerations in view of these findings.
Collapse
Affiliation(s)
- Lucy J Brooks
- Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, University College London Cancer Institute, London, UK.
| | - Holly Simpson Ragdale
- Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, University College London Cancer Institute, London, UK
| | - Ciaran Scott Hill
- Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, University College London Cancer Institute, London, UK; Department of Neurosurgery, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust (UCLH), London, UK
| | - Melanie Clements
- Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, University College London Cancer Institute, London, UK
| | - Simona Parrinello
- Samantha Dickson Brain Cancer Unit, Department of Cancer Biology, University College London Cancer Institute, London, UK.
| |
Collapse
|
18
|
Alexandris AS, Ryu J, Rajbhandari L, Harlan R, McKenney J, Wang Y, Aja S, Graham D, Venkatesan A, Koliatsos VE. Protective effects of NAMPT or MAPK inhibitors and NaR on Wallerian degeneration of mammalian axons. Neurobiol Dis 2022; 171:105808. [PMID: 35779777 PMCID: PMC10621467 DOI: 10.1016/j.nbd.2022.105808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/14/2022] [Accepted: 06/25/2022] [Indexed: 01/23/2023] Open
Abstract
Wallerian degeneration (WD) is a conserved axonal self-destruction program implicated in several neurological diseases. WD is driven by the degradation of the NAD+ synthesizing enzyme NMNAT2, the buildup of its substrate NMN, and the activation of the NAD+ degrading SARM1, eventually leading to axonal fragmentation. The regulation and amenability of these events to therapeutic interventions remain unclear. Here we explored pharmacological strategies that modulate NMN and NAD+ metabolism, namely the inhibition of the NMN-synthesizing enzyme NAMPT, activation of the nicotinic acid riboside (NaR) salvage pathway and inhibition of the NMNAT2-degrading DLK MAPK pathway in an axotomy model in vitro. Results show that NAMPT and DLK inhibition cause a significant but time-dependent delay of WD. These time-dependent effects are related to NMNAT2 degradation and changes in NMN and NAD+ levels. Supplementation of NAMPT inhibition with NaR has an enhanced effect that does not depend on timing of intervention and leads to robust protection up to 4 days. Additional DLK inhibition extends this even further to 6 days. Metabolite analyses reveal complex effects indicating that NAMPT and MAPK inhibition act by reducing NMN levels, ameliorating NAD+ loss and suppressing SARM1 activity. Finally, the axonal NAD+/NMN ratio is highly predictive of cADPR levels, extending previous cell-free evidence on the allosteric regulation of SARM1. Our findings establish a window of axon protection extending several hours following injury. Moreover, we show prolonged protection by mixed treatments combining MAPK and NAMPT inhibition that proceed via complex effects on NAD+ metabolism and inhibition of SARM1.
Collapse
Affiliation(s)
| | - Jiwon Ryu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Labchan Rajbhandari
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Harlan
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - James McKenney
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yiqing Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Aja
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - David Graham
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vassilis E Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Legarda SB, Lahti CE, McDermott D, Michas-Martin A. Use of Novel Concussion Protocol With Infralow Frequency Neuromodulation Demonstrates Significant Treatment Response in Patients With Persistent Postconcussion Symptoms, a Retrospective Study. Front Hum Neurosci 2022; 16:894758. [PMID: 35685335 PMCID: PMC9170890 DOI: 10.3389/fnhum.2022.894758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/20/2022] [Indexed: 01/28/2023] Open
Abstract
Introduction Concussion is a growing public health concern. No uniformly established therapy exists; neurofeedback studies report treatment value. We use infralow frequency neuromodulation (ILF) to remediate disabling neurological symptoms caused by traumatic brain injury (TBI) and noted improved outcomes with a novel concussion protocol. Postconcussion symptoms (PCS) and persistent postconcussion symptoms (PPCS; >3 months post head injury) are designated timelines for protracted neurological complaints following TBI. We performed a retrospective study to explore effectiveness of ILF in PCS/PPCS and investigated the value of using this concussion protocol. Method Patients with PCS/PPCS seen for their first neurology office visit or received their first neurofeedback session between 1 August 2018 and 31 January 2021 were entered. Outcomes were compared following treatment as usual (TAU) vs. TAU with ILF neurotherapy (TAU+ILF). The study cohort was limited to PPCS patients; the TAU+ILF group was restricted further to PPCS patients receiving at least 10 neurotherapy sessions. Within the TAU+ILF group, comparisons were made between those who trained at least 10 sessions using concussion protocol (TAU+ILF+CP) and those who trained for at least 10 sessions of ILF regardless of protocol (TAU+ILF-CP). Results Among our resultant PPCS cohort (n = 59) leading persistent neurological complaints were headache (67.8%), memory impairment (57.6%), and brain fog (50.8%). PPCS patients in TAU+ILF+CP (n = 25) demonstrated greater net (p = 0.004) and percent (p = 0.026) improvement of symptoms compared to PPCS subjects in TAU (n = 26). PPCS patients in TAU+ILF-CP (n = 8) trended toward significant symptom improvements compared to TAU, and TAU+ILF+CP trended toward greater efficacy than TAU+ILF-CP. Conclusion PPCS patients who received TAU+ILF+CP demonstrated significantly greater improvement as a group when compared to TAU. When used as an integrative modality to treatment as usual in managing patients with PPCS, ILF neuromodulation with use of concussion protocol provided significant symptom improvements.
Collapse
Affiliation(s)
- Stella B. Legarda
- Neurology, Montage Health, Montage Medical Group, Monterey, CA, United States
| | | | | | | |
Collapse
|
20
|
Radomski KL, Zi X, Lischka FW, Noble MD, Galdzicki Z, Armstrong RC. Acute axon damage and demyelination are mitigated by 4-aminopyridine (4-AP) therapy after experimental traumatic brain injury. Acta Neuropathol Commun 2022; 10:67. [PMID: 35501931 PMCID: PMC9059462 DOI: 10.1186/s40478-022-01366-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Damage to long axons in white matter tracts is a major pathology in closed head traumatic brain injury (TBI). Acute TBI treatments are needed that protect against axon damage and promote recovery of axon function to prevent long term symptoms and neurodegeneration. Our prior characterization of axon damage and demyelination after TBI led us to examine repurposing of 4-aminopyridine (4-AP), an FDA-approved inhibitor of voltage-gated potassium (Kv) channels. 4-AP is currently indicated to provide symptomatic relief for patients with chronic stage multiple sclerosis, which involves axon damage and demyelination. We tested clinically relevant dosage of 4-AP as an acute treatment for experimental TBI and found multiple benefits in corpus callosum axons. This randomized, controlled pre-clinical study focused on the first week after TBI, when axons are particularly vulnerable. 4-AP treatment initiated one day post-injury dramatically reduced axon damage detected by intra-axonal fluorescence accumulations in Thy1-YFP mice of both sexes. Detailed electron microscopy in C57BL/6 mice showed that 4-AP reduced pathological features of mitochondrial swelling, cytoskeletal disruption, and demyelination at 7 days post-injury. Furthermore, 4-AP improved the molecular organization of axon nodal regions by restoring disrupted paranode domains and reducing Kv1.2 channel dispersion. 4-AP treatment did not resolve deficits in action potential conduction across the corpus callosum, based on ex vivo electrophysiological recordings at 7 days post-TBI. Thus, this first study of 4-AP effects on axon damage in the acute period demonstrates a significant decrease in multiple pathological hallmarks of axon damage after experimental TBI.
Collapse
Affiliation(s)
- Kryslaine L. Radomski
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Xiaomei Zi
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Fritz W. Lischka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Mark D. Noble
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave, Box 633, Rochester, NY 14642 USA
| | - Zygmunt Galdzicki
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Regina C. Armstrong
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| |
Collapse
|
21
|
Gao L, Xue Q, Gong S, Li G, Tong W, Fan M, Chen X, Yin J, Song Y, Chen S, Huang J, Wang C, Dong Y. Structural and Functional Alterations of Substantia Nigra and Associations With Anxiety and Depressive Symptoms Following Traumatic Brain Injury. Front Neurol 2022; 13:719778. [PMID: 35449518 PMCID: PMC9017679 DOI: 10.3389/fneur.2022.719778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Backgrounds Although there are a certain number of studies dedicated to the disturbances of the dopaminergic system induced by traumatic brain injury (TBI), the associations of abnormal dopaminergic systems with post-traumatic anxiety and depressive disorders and their underlying mechanisms have not been clarified yet. In the midbrain, dopaminergic neurons are mainly situated in the substantia nigra (SN) and the ventral tegmental area (VTA). Thus, we selected SN and VTA as regions of interest and performed a seed-based global correlation to evaluate the altered functional connectivity throughout the dopaminergic system post-TBI. Methods Thirty-three individuals with TBI and 21 healthy controls were recruited in the study. Anxiety and depressive symptoms were examined by the Hospital Anxiety and Depression Scale. All MRI data were collected using a Siemens Prisma 3.0 Tesla MRI system. The volume of SN and the global functional connectivity of the SN and VTA were analyzed. Results In the present study, patients with TBI reported more anxiety and depressive symptoms. More importantly, some structural and functional alterations, such as smaller SN and reduced functional connectivity in the left SN, were seen in individuals with TBI. Patients with TBI had smaller substantia nigra on both right and left sides, and the left substantia nigra was relatively small in contrast with the right one. Among these findings, functional connectivity between left SN and left angular gyrus was positively associated with post-traumatic anxiety symptoms and negatively associated with depressive symptoms. Conclusions The TBI causes leftward lateralization of structural and functional alterations in the substantia nigra. An impaired mesocortical functional connectivity might be implicated in post-traumatic anxiety and depression.
Collapse
Affiliation(s)
- Liang Gao
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Xue
- Department of Neurosurgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Shun Gong
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Gaoyi Li
- Department of Neurosurgery, People's Hospital of Putuo District, Tongji University School of Medicine, Shanghai, China
| | - Wusong Tong
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Mingxia Fan
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China
| | - Xianzhen Chen
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Yin
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Song
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Songyu Chen
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingrong Huang
- Psychology Honors Program, University of California, San Diego, San Diego, CA, United States
| | - Chengbin Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Dong
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Tenth People's Hospital Clinical Medicine Scientific and Technical Innovation Park, Shanghai, China
| |
Collapse
|
22
|
Lu Q, Botchway BOA, Zhang Y, Jin T, Liu X. SARM1 can be a potential therapeutic target for spinal cord injury. Cell Mol Life Sci 2022; 79:161. [PMID: 35224705 PMCID: PMC11072485 DOI: 10.1007/s00018-022-04195-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/26/2022] [Accepted: 02/05/2022] [Indexed: 01/03/2023]
Abstract
Injury to the spinal cord is devastating. Studies have implicated Wallerian degeneration as the main cause of axonal destruction in the wake of spinal cord injury. Therefore, the suppression of Wallerian degeneration could be beneficial for spinal cord injury treatment. Sterile alpha and armadillo motif-containing protein 1 (SARM1) is a key modulator of Wallerian degeneration, and its impediment can improve spinal cord injury to a significant degree. In this report, we analyze the various signaling domains of SARM1, the recent findings on Wallerian degeneration and its relation to axonal insults, as well as its connection to SARM1, the mitogen-activated protein kinase (MAPK) signaling, and the survival factor, nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2). We then elaborate on the possible role of SARM1 in spinal cord injury and explicate how its obstruction could potentially alleviate the injury.
Collapse
Affiliation(s)
- Qicheng Lu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
| | - Tian Jin
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China.
| |
Collapse
|
23
|
MRI of Wallerian Degeneration in the Brainstem: A Pictorial Essay. J Belg Soc Radiol 2021; 105:58. [PMID: 34712899 PMCID: PMC8516011 DOI: 10.5334/jbsr.2585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/26/2021] [Indexed: 11/23/2022] Open
Abstract
Wallerian degeneration of the cerebral peduncle is a common MRI finding after cerebral injury. The degree of peduncular atrophy reflects the extent of damage in the corticospinal tract. The acute phase of Wallerian degeneration is visible with Diffusion-Weighted-Imaging. New investigation with Diffusion-Tensor-Imaging quantifies Wallerian degeneration in the subacute ischemic phase and is a good predictor for later functional recovery after stroke.
Collapse
|
24
|
Grovola MR, Paleologos N, Brown DP, Tran N, Wofford KL, Harris JP, Browne KD, Shewokis PA, Wolf JA, Cullen DK, Duda JE. Diverse changes in microglia morphology and axonal pathology during the course of 1 year after mild traumatic brain injury in pigs. Brain Pathol 2021; 31:e12953. [PMID: 33960556 PMCID: PMC8412066 DOI: 10.1111/bpa.12953] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/10/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Over 2.8 million people experience mild traumatic brain injury (TBI) in the United States each year, which may lead to long-term neurological dysfunction. The mechanical forces that are caused by TBI propagate through the brain to produce diffuse axonal injury (DAI) and trigger secondary neuroinflammatory cascades. The cascades may persist from acute to chronic time points after injury, altering the homeostasis of the brain. However, the relationship between the hallmark axonal pathology of diffuse TBI and potential changes in glial cell activation or morphology have not been established in a clinically relevant large animal model at chronic time points. In this study, we assessed the tissue from pigs subjected to rapid head rotation in the coronal plane to generate mild TBI. Neuropathological assessments for axonal pathology, microglial morphological changes, and astrocyte reactivity were conducted in specimens out to 1-year post-injury. We detected an increase in overall amyloid precursor protein pathology, as well as periventricular white matter and fimbria/fornix pathology after a single mild TBI. We did not detect the changes in corpus callosum integrity or astrocyte reactivity. However, detailed microglial skeletal analysis revealed changes in morphology, most notably increases in the number of microglial branches, junctions, and endpoints. These subtle changes were most evident in periventricular white matter and certain hippocampal subfields, and were observed out to 1-year post-injury in some cases. These ongoing morphological alterations suggest persistent change in neuroimmune homeostasis. Additional studies are needed to characterize the underlying molecular and neurophysiological alterations, as well as potential contributions to neurological deficits.
Collapse
Affiliation(s)
- Michael R. Grovola
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Nicholas Paleologos
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Daniel P. Brown
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Nathan Tran
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
| | - Kathryn L. Wofford
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - James P. Harris
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Kevin D. Browne
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Patricia A. Shewokis
- Department of Nutrition SciencesCollege of Nursing and Health ProfessionsDrexel UniversityPhiladelphiaPAUSA
- School of Biomedical Engineering, Science and Health SystemsDrexel UniversityPhiladelphiaPAUSA
| | - John A. Wolf
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - D. Kacy Cullen
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Center for Brain Injury & RepairDepartment of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPAUSA
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - John E. Duda
- Center for Neurotrauma, Neurodegeneration & RestorationCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Parkinson's Disease Research, Education and Clinical CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPAUSA
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| |
Collapse
|
25
|
Lacalle-Aurioles M, Cassel de Camps C, Zorca CE, Beitel LK, Durcan TM. Applying hiPSCs and Biomaterials Towards an Understanding and Treatment of Traumatic Brain Injury. Front Cell Neurosci 2020; 14:594304. [PMID: 33281561 PMCID: PMC7689345 DOI: 10.3389/fncel.2020.594304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and mortality in children and young adults and has a profound impact on the socio-economic wellbeing of patients and their families. Initially, brain damage is caused by mechanical stress-induced axonal injury and vascular dysfunction, which can include hemorrhage, blood-brain barrier disruption, and ischemia. Subsequent neuronal degeneration, chronic inflammation, demyelination, oxidative stress, and the spread of excitotoxicity can further aggravate disease pathology. Thus, TBI treatment requires prompt intervention to protect against neuronal and vascular degeneration. Rapid advances in the field of stem cells (SCs) have revolutionized the prospect of repairing brain function following TBI. However, more than that, SCs can contribute substantially to our knowledge of this multifaced pathology. Research, based on human induced pluripotent SCs (hiPSCs) can help decode the molecular pathways of degeneration and recovery of neuronal and glial function, which makes these cells valuable tools for drug screening. Additionally, experimental approaches that include hiPSC-derived engineered tissues (brain organoids and bio-printed constructs) and biomaterials represent a step forward for the field of regenerative medicine since they provide a more suitable microenvironment that enhances cell survival and grafting success. In this review, we highlight the important role of hiPSCs in better understanding the molecular pathways of TBI-related pathology and in developing novel therapeutic approaches, building on where we are at present. We summarize some of the most relevant findings for regenerative therapies using biomaterials and outline key challenges for TBI treatments that remain to be addressed.
Collapse
Affiliation(s)
- María Lacalle-Aurioles
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Camille Cassel de Camps
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Cornelia E Zorca
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Lenore K Beitel
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
26
|
Tomaiuolo F, Cerasa A, Lerch JP, Bivona U, Carlesimo GA, Ciurli P, Raffa G, Quattropani MC, Germanò A, Caltagirone C, Formisano R, Nigro S. Brain Neurodegeneration in the Chronic Stage of the Survivors from Severe Non-Missile Traumatic Brain Injury: A Voxel-Based Morphometry Within-Group at One versus Nine Years from a Head Injury. J Neurotrauma 2020; 38:283-290. [PMID: 32962533 DOI: 10.1089/neu.2020.7203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The long-term time course of neuropathological changes occurring in survivors from severe traumatic brain injury (TBI) remains uncertain. We investigated the brain morphometry and memory performance modifications within the same group of severe non-missile traumatic brain injury patients (nmTBI) after about ∼one year and at ∼ nine years from injury. Brain magnetic resonance imaging (MRI) measurements were performed with voxel-based morphometry (VBM) to determine specific changes in the gray matter (GM) and white matter (WM) and the overall gray matter volume modifications (GMV) and white matter volume modifications (WMV). Contemporarily, memory-tests were also administered. In comparison with healthy control subjects (HC), those with nmTBI showed a significant change and volume reduction in the GM and WM and also in the GMV and WMV after ∼one year; conversely, ∼nine years after injury, neurodegenerative changes spared the GM and GMV, but a prominent loss was detected in WMV and in WM sites, such as the superior longitudinal fasciculi, the body of the corpus callosum, the optic radiation, and the uncinate fasciculus. Memory performance at ∼one year in comparison with ∼nine years was stable with a subtle but significant trend toward recovery. These data demonstrate that patients with nmTBI undergo neurodegenerative processes during the chronic stage affecting mainly the cerebral WM rather than GM. Despite these anatomical brain parenchyma losses, memory performance tends to be stable or even slightly recovered. These results suggest possible correlations between progressive demyelinization and/or neuropsychiatric changes other than memory performance, and support possible treatments to prevent long-term WM degeneration of the examined nmTBI.
Collapse
Affiliation(s)
- Francesco Tomaiuolo
- Department of Clinical and Experimental Medicine and Department BIOMORF, University of Messina, Messina, Italy
| | - Antonio Cerasa
- IRIB, National Research Council, Cosenza, Italy, and S. Anna Institute and Research in Advanced Neurorehabilitation (RAN), Crotone, Italy
| | - Jason P Lerch
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, The University of Oxford, Oxford, United Kingdom
| | | | - Giovanni Augusto Carlesimo
- IRCCS Fondazione 'Santa Lucia', Rome, Italy.,Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | | | - Giovanni Raffa
- Division of Neurosurgery, Department BIOMORF, University of Messina, Messina, Italy
| | - Marina Catena Quattropani
- Department of Clinical and Experimental Medicine and Department BIOMORF, University of Messina, Messina, Italy
| | - Antonino Germanò
- Division of Neurosurgery, Department BIOMORF, University of Messina, Messina, Italy
| | | | | | - Salvatore Nigro
- Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy
| |
Collapse
|
27
|
Bartnik-Olson B, Holshouser B, Ghosh N, Oyoyo UE, Nichols JG, Pivonka-Jones J, Tong K, Ashwal S. Evolving White Matter Injury following Pediatric Traumatic Brain Injury. J Neurotrauma 2020; 38:111-121. [PMID: 32515269 DOI: 10.1089/neu.2019.6574] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
This study is unique in that it examines the evolution of white matter injury very early and at 12 months post-injury in pediatric patients following traumatic brain injury (TBI). Diffusion tensor imaging (DTI) was acquired at two time-points: acutely at 6-17 days and 12 months following a complicated mild (cMild)/moderate (mod) or severe TBI. Regional measures of anisotropy and diffusivity were compared between TBI groups and against a group of age-matched healthy controls and used to predict performance on measures of attention, memory, and intellectual functioning at 12-months post-injury. Analysis of the acute DTI data using tract based spatial statistics revealed a small number of regional decreases in fractional anisotropy (FA) in both the cMild/mod and severe TBI groups compared with controls. These changes were observed in the occipital white matter, anterior limb of the internal capsule (ALIC)/basal ganglia, and corpus callosum. The severe TBI group showed regional differences in axial diffusivity (AD) in the brainstem and corpus callosum that were not seen in the cMild/mod TBI group. By 12-months, widespread decreases in FA and increases in apparent diffusion coefficient (ADC) and radial diffusivity (RD) were observed in both TBI groups compared with controls, with the overall number of regions with abnormal DTI metrics increasing over time. The early changes in regional DTI metrics were associated with 12-month performance IQ scores. These findings suggest that there may be regional differences in the brain's reparative processes or that mechanisms associated with the brain's plasticity to recover may also be region based.
Collapse
Affiliation(s)
- Brenda Bartnik-Olson
- Department of Radiology, Loma Linda University Health, Loma Linda, California, USA
| | - Barbara Holshouser
- Department of Radiology, Loma Linda University Health, Loma Linda, California, USA
| | - Nirmalya Ghosh
- Department of Pediatrics, Loma Linda University Health, Loma Linda, California, USA
| | - Udochukwu E Oyoyo
- Department of Radiology, Loma Linda University Health, Loma Linda, California, USA
| | - Joy G Nichols
- Department of Pediatrics, Loma Linda University Health, Loma Linda, California, USA
| | - Jamie Pivonka-Jones
- Department of Pediatrics, Loma Linda University Health, Loma Linda, California, USA
| | - Karen Tong
- Department of Radiology, Loma Linda University Health, Loma Linda, California, USA
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University Health, Loma Linda, California, USA
| |
Collapse
|
28
|
Abstract
Traumatic brain injury is a calamity of various causes, pathologies, and extremely varied and often complex clinical presentations. Because of its predilection for brain systems underlying cognitive and complex behavioral operations, it may cause chronic and severe psychiatric illness that requires expert management. This is more so for the modern epidemic of athletic and military brain injuries which are dominated by psychiatric symptoms. Past medical, including psychiatric, history, and comorbidities are important and relevant for formulation and management. Traumatic brain injury is a model for other neuropsychiatric disorders and may serve as an incubator of new ideas for neurodegenerative disease.
Collapse
Affiliation(s)
- Vassilis E Koliatsos
- Department of Pathology (Neuropathology), Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Neuropsychiatry Program, Sheppard Pratt Health System, Baltimore, MD, USA.
| | - Vani Rao
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Abstract
Microglia dynamically interact with neurons influencing the development, structure, and function of neuronal networks. Recent studies suggest microglia may also influence neuronal activity by physically interacting with axonal domains responsible for action potential initiation and propagation. However, the nature of these microglial process interactions is not well understood. Microglial-axonal contacts are present early in development and persist through adulthood, implicating microglial interactions in the regulation of axonal integrity in both the developing and mature central nervous system. Moreover, changes in microglial-axonal contact have been described in disease states such as multiple sclerosis (MS) and traumatic brain injury (TBI). Depending on the disease state, there are increased associations with specific axonal segments. In MS, there is enhanced contact with the axon initial segment and node of Ranvier, while, in TBI, microglia alter interactions with axons at the site of injury, as well as at the axon initial segment. In this article, we review the interactions of microglial processes with axonal segments, analyzing their associations with various axonal domains and how these interactions may differ between MS and TBI. Furthermore, we discuss potential functional consequences and molecular mechanisms of these interactions and how these may differ among various types of microglial-axonal interactions.
Collapse
Affiliation(s)
- Savannah D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Audrey D Lafrenaye
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
30
|
Editorial: The voluminous axon as an organizing principle for traumatic brain injury therapeutics: novel molecular, metabolic, and circuit strategies. Curr Opin Neurol 2019; 32:783-785. [PMID: 31609737 DOI: 10.1097/wco.0000000000000757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|