Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ann Neurol ; 96(2): 365-377, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38845484

ABSTRACT

OBJECTIVE: The long-term consequences of traumatic brain injury (TBI) on brain structure remain uncertain. Given evidence that a single significant brain injury event increases the risk of dementia, brain-age estimation could provide a novel and efficient indexing of the long-term consequences of TBI. Brain-age procedures use predictive modeling to calculate brain-age scores for an individual using structural magnetic resonance imaging (MRI) data. Complicated mild, moderate, and severe TBI (cmsTBI) is associated with a higher predicted age difference (PAD), but the progression of PAD over time remains unclear. We sought to examine whether PAD increases as a function of time since injury (TSI) and if injury severity and sex interacted to influence this progression. METHODS: Through the ENIGMA Adult Moderate and Severe (AMS)-TBI working group, we examine the largest TBI sample to date (n = 343), along with controls, for a total sample size of n = 540, to replicate and extend prior findings in the study of TBI brain age. Cross-sectional T1w-MRI data were aggregated across 7 cohorts, and brain age was established using a similar brain age algorithm to prior work in TBI. RESULTS: Findings show that PAD widens with longer TSI, and there was evidence for differences between sexes in PAD, with men showing more advanced brain age. We did not find strong evidence supporting a link between PAD and cognitive performance. INTERPRETATION: This work provides evidence that changes in brain structure after cmsTBI are dynamic, with an initial period of change, followed by relative stability in brain morphometry, eventually leading to further changes in the decades after a single cmsTBI. ANN NEUROL 2024;96:365-377.


Subject(s)
Brain Injuries, Traumatic , Magnetic Resonance Imaging , Humans , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/complications , Male , Female , Adult , Middle Aged , Cohort Studies , Brain/diagnostic imaging , Brain/pathology , Aged , Aging/pathology , Aging, Premature/diagnostic imaging , Aging, Premature/pathology
2.
Neurobiol Dis ; 171: 105808, 2022 09.
Article in English | MEDLINE | ID: mdl-35779777

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.


Subject(s)
Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase , Wallerian Degeneration , Animals , Armadillo Domain Proteins/metabolism , Axons/pathology , Cytoskeletal Proteins/metabolism , Humans , Mammals/metabolism , NAD/metabolism , Nerve Degeneration/pathology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Protein Kinase Inhibitors , Wallerian Degeneration/metabolism
3.
Int J Mol Sci ; 23(13)2022 Jul 02.
Article in English | MEDLINE | ID: mdl-35806394

ABSTRACT

White matter pathology is common across a wide spectrum of neurological diseases. Characterizing this pathology is important for both a mechanistic understanding of neurological diseases as well as for the development of neuroimaging biomarkers. Although axonal calibers can vary by orders of magnitude, they are tightly regulated and related to neuronal function, and changes in axon calibers have been reported in several diseases and their models. In this study, we utilize the impact acceleration model of traumatic brain injury (IA-TBI) to assess early and late changes in the axon diameter distribution (ADD) of the mouse corticospinal tract using Airyscan and electron microscopy. We find that axon calibers follow a lognormal distribution whose parameters significantly change after injury. While IA-TBI leads to 30% loss of corticospinal axons by day 7 with a bias for larger axons, at 21 days after injury we find a significant redistribution of axon frequencies that is driven by a reduction in large-caliber axons in the absence of detectable degeneration. We postulate that changes in ADD features may reflect a functional adaptation of injured neural systems. Moreover, we find that ADD features offer an accurate way to discriminate between injured and non-injured mice. Exploring injury-related ADD signatures by histology or new emerging neuroimaging modalities may offer a more nuanced and comprehensive way to characterize white matter pathology and may also have the potential to generate novel biomarkers of injury.


Subject(s)
Brain Injuries, Traumatic , White Matter , Animals , Axons/pathology , Brain Injuries, Traumatic/pathology , Mice , Mice, Inbred Strains , Pyramidal Tracts/pathology , White Matter/pathology
4.
J Neurosci ; 38(16): 4031-4047, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29567804

ABSTRACT

Traumatic axonal injury (TAI) is a common neuropathology in traumatic brain injury and is featured by primary injury to axons. Here, we generated TAI with impact acceleration of the head in male Thy1-eYFP-H transgenic mice in which specific populations of neurons and their axons are labeled with yellow fluorescent protein. This model results in axonal lesions in multiple axonal tracts along with blood-brain barrier disruption and neuroinflammation. The corticospinal tract, a prototypical long tract, is severely affected and is the focus of this study. Using optimized CLARITY at single-axon resolution, we visualized the entire corticospinal tract volume from the pons to the cervical spinal cord in 3D and counted the total number of axonal lesions and their progression over time. Our results divulged the presence of progressive traumatic axonopathy that was maximal at the pyramidal decussation. The perikarya of injured corticospinal neurons atrophied, but there was no evidence of neuronal cell death. We also used CLARITY at single-axon resolution to explore the role of the NMNAT2-SARM1 axonal self-destruction pathway in traumatic axonopathy. When we interfered with this pathway by genetically ablating SARM1 or by pharmacological strategies designed to increase levels of Nicotinamide (Nam), a feedback inhibitor of SARM1, we found a significant reduction in the number of axonal lesions early after injury. Our findings show that high-resolution neuroanatomical strategies reveal important features of TAI with biological implications, especially the progressive axonopathic nature of TAI and the role of the NMNAT2-SARM1 pathway in the early stages of axonopathy.SIGNIFICANCE STATEMENT In the first systematic application of novel high-resolution neuroanatomical tools in neuropathology, we combined CLARITY with 2-photon microscopy, optimized for detection of single axonal lesions, to reconstruct the injured mouse brainstem in a model of traumatic axonal injury (TAI) that is a common pathology associated with traumatic brain injury. The 3D reconstruction of the corticospinal tract at single-axon resolution allowed for a more advanced level of qualitative and quantitative understanding of TAI. Using this model, we showed that TAI is an axonopathy with a prominent role of the NMNAT2-SARM1 molecular pathway, that is also implicated in peripheral neuropathy. Our results indicate that high-resolution anatomical models of TAI afford a level of detail and sensitivity that is ideal for testing novel molecular and biomechanical hypotheses.


Subject(s)
Axons/pathology , Brain Injuries, Traumatic/pathology , Microscopy, Fluorescence, Multiphoton/methods , Pyramidal Tracts/pathology , Animals , Armadillo Domain Proteins/metabolism , Axons/metabolism , Brain Injuries, Traumatic/diagnostic imaging , Cytoskeletal Proteins/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton/standards , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Sensitivity and Specificity , Single-Cell Analysis/methods , Single-Cell Analysis/standards
5.
Curr Opin Neurol ; 32(6): 786-795, 2019 12.
Article in English | MEDLINE | ID: mdl-31633494

ABSTRACT

PURPOSE OF REVIEW: Diffuse or traumatic axonal injury is one of the principal pathologies encountered in traumatic brain injury (TBI) and the resulting axonal loss, disconnection, and brain atrophy contribute significantly to clinical morbidity and disability. The seminal discovery of the slow Wallerian degeneration mice (Wld) in which transected axons do not degenerate but survive and function independently for weeks has transformed concepts on axonal biology and raised hopes that axonopathies may be amenable to specific therapeutic interventions. Here we review mechanisms of axonal degeneration and also describe how these mechanisms may inform biological therapies of traumatic axonopathy in the context of TBI. RECENT FINDINGS: In the last decade, SARM1 [sterile a and Toll/interleukin-1 receptor (TIR) motif containing 1] and the DLK (dual leucine zipper bearing kinase) and LZK (leucine zipper kinase) MAPK (mitogen-activated protein kinases) cascade have been established as the key drivers of Wallerian degeneration, a complex program of axonal self-destruction which is activated by a wide range of injurious insults, including insults that may otherwise leave axons structurally robust and potentially salvageable. Detailed studies on animal models and postmortem human brains indicate that this type of partial disruption is the main initial pathology in traumatic axonopathy. At the same time, the molecular dissection of Wallerian degeneration has revealed that the decision that commits axons to degeneration is temporally separated from the time of injury, a window that allows potentially effective pharmacological interventions. SUMMARY: Molecular signals initiating and triggering Wallerian degeneration appear to be playing an important role in traumatic axonopathy and recent advances in understanding their nature and significance is opening up new therapeutic opportunities for TBI.


Subject(s)
Axons , Brain Injuries, Diffuse , Brain Injuries, Traumatic , Wallerian Degeneration , Animals , Axons/metabolism , Axons/pathology , Brain Injuries, Diffuse/metabolism , Brain Injuries, Diffuse/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Humans , Wallerian Degeneration/drug therapy , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology
6.
Hum Mol Genet ; 25(16): 3515-3523, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27378696

ABSTRACT

Multiple missense mutations in Leucine-rich repeat kinase 2 (LRRK2) have been linked to Parkinson's disease (PD), the most common degenerative movement disorder. LRRK2 is expressed by both neurons and microglia, the residential immune cells in the brain. Increasing evidence supports a role of LRRK2 in modulating microglial activity, of which Lrrk2-null rodent microglia display less inflammatory response to endotoxin lipopolysaccharide (LPS). The underlying molecular mechanism, however, remains elusive. Chemokine (C-X3-C) receptor 1 (CX3CR1), predominantly expressed by microglia, suppresses microglial inflammation while promotes migration. Using whole-genome microarray screening, we found that Cx3cr1 mRNA levels were substantially higher in microglia derived from Lrrk2 knockout (Lrrk2-/-) mice. The total and cell surface levels of CX3CR1 proteins were also remarkably increased. In correlation with the enhanced CX3CR1 expression, Lrrk2-null microglia migrated faster and travelled longer distance toward the source of fractalkine (CX3CL1), an endogenous ligand of CX3CR1. To investigate the impact of CX3CR1 elevation in vivo, we compared LPS-induced inflammation in the striatum of Lrrk2-/- knockout mice with Cx3cr1 heterozygous and homozygous knockout background. We found that a complete loss of Cx3cr1 restored the responsiveness of Lrrk2-/- microglia to LPS stimulation. In conclusion, our findings reveal a previously unknown regulatory role for LRRK2 in CX3CR1 signalling and suggest that an increase of CX3CR1 activity contributes to the attenuated inflammatory responses in Lrrk2-null microglia.


Subject(s)
Inflammation/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease/genetics , Receptors, Chemokine/genetics , Animals , CX3C Chemokine Receptor 1 , Corpus Striatum/metabolism , Corpus Striatum/pathology , Humans , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/administration & dosage , Macrophage Activation/drug effects , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Parkinson Disease/pathology , Receptors, Chemokine/biosynthesis , Signal Transduction/genetics
7.
Brain Inj ; 31(9): 1168-1176, 2017.
Article in English | MEDLINE | ID: mdl-28981339

ABSTRACT

As a follow-up to the 2008 state-of-the-art (SOTA) conference on traumatic brain injuries (TBIs), the 2015 event organized by the United States Department of Veterans Affairs (VA) Office of Research and Development (ORD) analysed the knowledge gained over the last 7 years as it relates to basic scientific methods, experimental findings, diagnosis, therapy, and rehabilitation of TBIs and blast-induced neurotraumas (BINTs). The current article summarizes the discussions and recommendations of the scientific panel attending the Preclinical Modeling and Therapeutic Development Workshop of the conference, with special emphasis on factors slowing research progress and recommendations for ways of addressing the most significant pitfalls.


Subject(s)
Blast Injuries/epidemiology , Brain Injuries, Traumatic/epidemiology , Disease Models, Animal , Military Personnel , United States Department of Veterans Affairs/trends , Animals , Blast Injuries/psychology , Blast Injuries/therapy , Brain Injuries, Traumatic/psychology , Brain Injuries, Traumatic/therapy , Forecasting , Humans , Military Personnel/psychology , United States/epidemiology
8.
Int J Geriatr Psychiatry ; 31(12): 1301-1311, 2016 12.
Article in English | MEDLINE | ID: mdl-26876501

ABSTRACT

OBJECTIVE: Depression may be a prodrome to Alzheimer's disease (AD). We assessed whether AD neuropathology is associated with depression in mild cognitive impairment (MCI) and mild dementia (dAD). METHODS: All clinical and neuropathological data for this study came from the National Alzheimer's Coordinating Center (NACC). Healthy control (HC, n = 120), MCI (n = 77), and mild dAD (n = 93) patients who underwent brain autopsy were included. In regression models with Geriatric Depression Scale (GDS) as the outcome, neuritic plaque (NP) score or Braak Stages of neurofibrillary (NF) pathology were covariates. RESULTS: GDS was not associated with cognitive status, NP score, Braak Stages, or their interaction. In both models, a history of TIAs, depression within the last 2 years, current benzodiazepine use, and greater severity of neuropsychiatric symptoms were associated with greater depression. In the Braak Stages model, less education was another significant predictor. CONCLUSIONS: Depression in early AD appears to be independent of NP and NF pathology. Studies are needed to investigate other mechanisms that may be responsible for depression in MCI and dAD.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/psychology , Brain/pathology , Depressive Disorder/pathology , Depressive Disorder/psychology , Aged , Aged, 80 and over , Autopsy , Case-Control Studies , Cognitive Dysfunction/psychology , Dementia/psychology , Female , Humans , Male , Neuropsychological Tests , Plaque, Amyloid/pathology , Psychiatric Status Rating Scales , Regression Analysis
9.
Antioxid Redox Signal ; 39(16-18): 1167-1184, 2023 12.
Article in English | MEDLINE | ID: mdl-37503611

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.


Subject(s)
Neurodegenerative Diseases , Peripheral Nervous System Diseases , Humans , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , NAD/metabolism , Peripheral Nervous System Diseases/metabolism , Axons/metabolism , Neurodegenerative Diseases/metabolism
10.
Exp Neurol ; 359: 114252, 2023 01.
Article in English | MEDLINE | ID: mdl-36244414

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.


Subject(s)
Brain Injuries, Traumatic , Wallerian Degeneration , Animals , Mice , Wallerian Degeneration/etiology , Axons/pathology , Myelin Sheath/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Acceleration , Cytoskeletal Proteins/genetics , Armadillo Domain Proteins/genetics
11.
J Neurotrauma ; 40(15-16): 1743-1761, 2023 08.
Article in English | MEDLINE | ID: mdl-36680758

ABSTRACT

Traumatic axonal injury (TAI), thought to be caused by rotational acceleration of the head, is a prevalent neuropathology in traumatic brain injury (TBI). TAI in the optic nerve is a common finding in multiple blunt-force TBI models and hence a great model to study mechanisms and treatments for TAI, especially in view of the compartmentalized anatomy of the visual system. We have previously shown that the somata and the proximal, but not distal, axons of retinal ganglion cells (RGC) respond to DLK/LZK blockade after impact acceleration of the head (IA-TBI). Here, we explored the role of the sterile alpha and TIR-motif containing 1 (SARM1), the key driver of Wallerian degeneration (WD), in the progressive breakdown of distal and proximal segments of the optic nerve following IA-TBI with high-resolution morphological and classical neuropathological approaches. Wild type and Sarm1 knockout (KO) mice received IA-TBI or sham injury and were allowed to survive for 3, 7, 14, and 21 days. Ultrastructural and microscopic analyses revealed that TAI in the optic nerve is characterized by variable involvement of individual axons, ranging from apparent early disconnection of a subpopulation of axons to a range of ongoing axonal and myelin perturbations. Traumatic axonal injury resulted in the degeneration of a population of axons distal and proximal to the injury, along with retrograde death of a subpopulation of RGCs. Quantitative analyses on proximal and distal axons and RGC somata revealed that different neuronal domains exhibit differential vulnerability, with distal axon segments showing more severe degeneration compared with proximal segments and RGC somata. Importantly, we found that Sarm1 KO had a profound effect in the distal optic nerve by suppressing axonal degeneration by up to 50% in the first 2 weeks after IA-TBI, with a continued but lower effect at 3 weeks, while also suppressing microglial activation. Sarm1 KO had no evident effect on the initial traumatic disconnection and did not ameliorate the proximal optic axonopathy or the subsequent attrition of RGCs, indicating that the fate of different axonal segments in the course of TAI may depend on distinct molecular programs within axons.


Subject(s)
Axons , Brain Injuries, Traumatic , Mice , Animals , Axons/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Brain Injuries, Traumatic/pathology , Optic Nerve/pathology , Mice, Knockout , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism
12.
Neurobiol Dis ; 41(2): 538-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21074615

ABSTRACT

Current experimental models of blast injuries used to study blast-induced neurotrauma (BINT) vary widely, which makes the comparison of the experimental results extremely challenging. Most of the blast injury models replicate the ideal Friedländer type of blast wave, without the capability to generate blast signatures with multiple shock fronts and refraction waves as seen in real-life conditions; this significantly reduces their clinical and military relevance. Here, we describe the pathophysiological consequences of graded blast injuries and BINT generated by a newly developed, highly controlled, and reproducible model using a modular, multi-chamber shock tube capable of tailoring pressure wave signatures and reproducing complex shock wave signatures seen in theater. While functional deficits due to blast exposure represent the principal health problem for today's warfighters, the majority of available blast models induces tissue destruction rather than mimic functional deficits. Thus, the main goal of our model is to reliably reproduce long-term neurological impairments caused by blast. Physiological parameters, functional (motor, cognitive, and behavioral) outcomes, and underlying molecular mechanisms involved in inflammation measured in the brain over the 30 day post-blast period showed this model is capable of reproducing major neurological changes of clinical BINT.


Subject(s)
Blast Injuries/diagnosis , Blast Injuries/pathology , Brain Injuries/diagnosis , Brain Injuries/pathology , Pressure/adverse effects , Animals , Atmosphere Exposure Chambers/adverse effects , Atmosphere Exposure Chambers/standards , Atmospheric Pressure , Blast Injuries/physiopathology , Brain Injuries/physiopathology , Disease Models, Animal , Environment, Controlled , Male , Mice , Mice, Inbred C57BL
13.
Brain Imaging Behav ; 15(2): 526-554, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32797398

ABSTRACT

The global burden of mortality and morbidity caused by traumatic brain injury (TBI) is significant, and the heterogeneity of TBI patients and the relatively small sample sizes of most current neuroimaging studies is a major challenge for scientific advances and clinical translation. The ENIGMA (Enhancing NeuroImaging Genetics through Meta-Analysis) Adult moderate/severe TBI (AMS-TBI) working group aims to be a driving force for new discoveries in AMS-TBI by providing researchers world-wide with an effective framework and platform for large-scale cross-border collaboration and data sharing. Based on the principles of transparency, rigor, reproducibility and collaboration, we will facilitate the development and dissemination of multiscale and big data analysis pipelines for harmonized analyses in AMS-TBI using structural and functional neuroimaging in combination with non-imaging biomarkers, genetics, as well as clinical and behavioral measures. Ultimately, we will offer investigators an unprecedented opportunity to test important hypotheses about recovery and morbidity in AMS-TBI by taking advantage of our robust methods for large-scale neuroimaging data analysis. In this consensus statement we outline the working group's short-term, intermediate, and long-term goals.


Subject(s)
Brain Injuries, Traumatic , Magnetic Resonance Imaging , Adult , Brain/diagnostic imaging , Humans , Neuroimaging , Reproducibility of Results
14.
Stem Cells ; 27(10): 2414-26, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19609935

ABSTRACT

Stem cell grafts have been advocated as experimental treatments for neurological diseases by virtue of their ability to offer trophic support for injured neurons and, theoretically, to replace dead neurons. Human embryonic stem cells (HESCs) are a rich source of neural precursors (NPs) for grafting, but have been questioned for their tendency to form tumors. Here we studied the ability of HESC-derived NP grafts optimized for cell number and differentiation stage prior to transplantation, to survive and stably differentiate and integrate in the basal forebrain (neostriatum) of young adult nude rats over long periods of time (6 months). NPs were derived from adherent monolayer cultures of HESCs exposed to noggin. After transplantation, NPs showed a drastic reduction in mitotic activity and an avid differentiation into neurons that projected via major white matter tracts to a variety of forebrain targets. A third of NP-derived neurons expressed the basal forebrain-neostriatal marker dopamine-regulated and cyclic AMP-regulated phosphoprotein. Graft-derived neurons formed mature synapses with host postsynaptic structures, including dendrite shafts and spines. NPs inoculated in white matter tracts showed a tendency toward glial (primarily astrocytic) differentiation, whereas NPs inoculated in the ventricular epithelium persisted as nestin(+) precursors. Our findings demonstrate the long-term ability of noggin-derived human NPs to structurally integrate tumor-free into the mature mammalian forebrain, while maintaining some cell fate plasticity that is strongly influenced by particular central nervous system (CNS) niches.


Subject(s)
Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Neostriatum/physiology , Stem Cell Transplantation/methods , Stem Cells/physiology , Transplantation, Heterologous/physiology , Animals , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Survival/physiology , Embryonic Stem Cells/cytology , Graft Survival/physiology , Growth Cones/physiology , Growth Cones/ultrastructure , Humans , Male , Mice , Mice, Inbred ICR , Neostriatum/cytology , Neostriatum/surgery , Neural Pathways/cytology , Neural Pathways/physiology , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Phosphoproteins/metabolism , Rats , Rats, Nude , Stem Cells/cytology , Synapses/ultrastructure
16.
Psychiatr Clin North Am ; 43(2): 291-304, 2020 06.
Article in English | MEDLINE | ID: mdl-32439023

ABSTRACT

This article reviews some of the recent discoveries about how neurobiological processes contribute to the understanding and treatment of substance use disorders. Particular focus is given to cannabis, opioids, and designer drugs. Important areas addressed include triggers and cravings, the central roles of dopamine and stress, and the endocannabinoid system. Clinical relevance of these findings for withdrawal management and relapse prevention is discussed. Also highlighted are issues related to the opioid epidemic and consequences both of continuing federal prohibition of cannabis as well as its state-by-state relaxation.


Subject(s)
Opioid-Related Disorders/therapy , Substance-Related Disorders/therapy , Analgesics, Opioid , Craving , Humans , Substance Withdrawal Syndrome/drug therapy
17.
Psychiatr Clin North Am ; 43(2): 305-330, 2020 06.
Article in English | MEDLINE | ID: mdl-32439024

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.


Subject(s)
Brain Injuries, Traumatic/psychology , Blast Injuries/psychology , Brain Contusion/psychology , Diffuse Axonal Injury/psychology , Humans , Military Personnel , Stress Disorders, Post-Traumatic/psychology
18.
Psychiatr Clin North Am ; 43(2): 213-227, 2020 06.
Article in English | MEDLINE | ID: mdl-32439018

ABSTRACT

Neuropsychiatry is an integrative discipline defined by its history, its preferred patients, and its theoretic framework. Dealing with human behavior needs to consider the brain, but such consideration should avoid oversimplification: neurologic understanding is not essential, necessary, or desirable in all conditions encountered in clinical psychiatry. Neuropsychiatric theory is founded on discoveries in the areas of synaptic plasticity and cortical/limbic anatomy (bottom-up), but also evolutionary biology and anthropology (top-down). Going forward, we need to synthesize vital information, distinguish the essential from the trivial or tenuous, and remain open to dialogue with allied disciplines, our patients, and our students.


Subject(s)
Mental Disorders/pathology , Neuropsychiatry , History, 19th Century , History, 20th Century , Humans
19.
PLoS One ; 14(11): e0224846, 2019.
Article in English | MEDLINE | ID: mdl-31710637

ABSTRACT

Optogenetically engineered human neural progenitors (hNPs) are viewed as promising tools in regenerative neuroscience because they allow the testing of the ability of hNPs to integrate within nervous system of an appropriate host not only structurally, but also functionally based on the responses of their differentiated progenies to light. Here, we transduced H9 embryonic stem cell-derived hNPs with a lentivirus harboring human channelrhodopsin (hChR2) and differentiated them into a forebrain lineage. We extensively characterized the fate and optogenetic functionality of hChR2-hNPs in vitro with electrophysiology and immunocytochemistry. We also explored whether the in vivo phenotype of ChR2-hNPs conforms to in vitro observations by grafting them into the frontal neocortex of rodents and analyzing their survival and neuronal differentiation. Human ChR2-hNPs acquired neuronal phenotypes (TUJ1, MAP2, SMI-312, and synapsin 1 immunoreactivity) in vitro after an average of 70 days of coculturing with CD1 astrocytes and progressively displayed both inhibitory and excitatory neurotransmitter signatures by immunocytochemistry and whole-cell patch clamp recording. Three months after transplantation into motor cortex of naïve or injured mice, 60-70% of hChR2-hNPs at the transplantation site expressed TUJ1 and had neuronal cytologies, whereas 60% of cells also expressed ChR2. Transplant-derived neurons extended axons through major commissural and descending tracts and issued synaptophysin+ terminals in the claustrum, endopiriform area, and corresponding insular and piriform cortices. There was no apparent difference in engraftment, differentiation, or connectivity patterns between injured and sham subjects. Same trends were observed in a second rodent host, i.e. rat, where we employed longer survival times and found that the majority of grafted hChR2-hNPs differentiated into GABAergic neurons that established dense terminal fields and innervated mostly dendritic profiles in host cortical neurons. In physiological experiments, human ChR2+ neurons in culture generated spontaneous action potentials (APs) 100-170 days into differentiation and their firing activity was consistently driven by optical stimulation. Stimulation generated glutamatergic and GABAergic postsynaptic activity in neighboring ChR2- cells, evidence that hChR2-hNP-derived neurons had established functional synaptic connections with other neurons in culture. Light stimulation of hChR2-hNP transplants in vivo generated complicated results, in part because of the variable response of the transplants themselves. Our findings show that we can successfully derive hNPs with optogenetic properties that are fully transferrable to their differentiated neuronal progenies. We also show that these progenies have substantial neurotransmitter plasticity in vitro, whereas in vivo they mostly differentiate into inhibitory GABAergic neurons. Furthermore, neurons derived from hNPs have the capacity of establishing functional synapses with postsynaptic neurons in vitro, but this outcome is technically challenging to explore in vivo. We propose that optogenetically endowed hNPs hold great promise as tools to explore de novo circuit formation in the brain and, in the future, perhaps launch a new generation of neuromodulatory therapies.


Subject(s)
Human Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Optogenetics , Animals , Astrocytes/cytology , Astrocytes/radiation effects , Axons/metabolism , Axons/radiation effects , Cell Differentiation/radiation effects , Cell Lineage/radiation effects , Cell Survival/radiation effects , Channelrhodopsins/metabolism , Human Embryonic Stem Cells/radiation effects , Humans , Lentivirus/metabolism , Light , Mice, Nude , Motor Cortex/metabolism , Neural Stem Cells/radiation effects , Neuronal Plasticity/radiation effects , Neurons/radiation effects , Neurotransmitter Agents/metabolism , Phenotype , Photic Stimulation , Rats, Nude , Synaptic Transmission/radiation effects
20.
Mol Neurodegener ; 14(1): 44, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31775817

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a major cause of CNS neurodegeneration and has no disease-altering therapies. It is commonly associated with a specific type of biomechanical disruption of the axon called traumatic axonal injury (TAI), which often leads to axonal and sometimes perikaryal degeneration of CNS neurons. We have previously used genome-scale, arrayed RNA interference-based screens in primary mouse retinal ganglion cells (RGCs) to identify a pair of related kinases, dual leucine zipper kinase (DLK) and leucine zipper kinase (LZK) that are key mediators of cell death in response to simple axotomy. Moreover, we showed that DLK and LZK are the major upstream triggers for JUN N-terminal kinase (JNK) signaling following total axonal transection. However, the degree to which DLK/LZK are involved in TAI/TBI is unknown. METHODS: Here we used the impact acceleration (IA) model of diffuse TBI, which produces TAI in the visual system, and complementary genetic and pharmacologic approaches to disrupt DLK and LZK, and explored whether DLK and LZK play a role in RGC perikaryal and axonal degeneration in response to TAI. RESULTS: Our findings show that the IA model activates DLK/JNK/JUN signaling but, in contrast to axotomy, many RGCs are able to recover from the injury and terminate the activation of the pathway. Moreover, while DLK disruption is sufficient to suppress JUN phosphorylation, combined DLK and LZK inhibition is required to prevent RGC cell death. Finally, we show that the FDA-approved protein kinase inhibitor, sunitinib, which has activity against DLK and LZK, is able to produce similar increases in RGC survival. CONCLUSION: The mitogen-activated kinase kinase kinases (MAP3Ks), DLK and LZK, participate in cell death signaling of CNS neurons in response to TBI. Moreover, sustained pharmacologic inhibition of DLK is neuroprotective, an effect creating an opportunity to potentially translate these findings to patients with TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Cell Survival/physiology , MAP Kinase Kinase Kinases/metabolism , Neurons/metabolism , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Leucine Zippers/drug effects , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Neurons/drug effects , Protein Kinase Inhibitors/pharmacology , Retinal Ganglion Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL