Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell ; 184(10): 2715-2732.e23, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33852912

ABSTRACT

Traumatic brain injury (TBI) is the largest non-genetic, non-aging related risk factor for Alzheimer's disease (AD). We report here that TBI induces tau acetylation (ac-tau) at sites acetylated also in human AD brain. This is mediated by S-nitrosylated-GAPDH, which simultaneously inactivates Sirtuin1 deacetylase and activates p300/CBP acetyltransferase, increasing neuronal ac-tau. Subsequent tau mislocalization causes neurodegeneration and neurobehavioral impairment, and ac-tau accumulates in the blood. Blocking GAPDH S-nitrosylation, inhibiting p300/CBP, or stimulating Sirtuin1 all protect mice from neurodegeneration, neurobehavioral impairment, and blood and brain accumulation of ac-tau after TBI. Ac-tau is thus a therapeutic target and potential blood biomarker of TBI that may represent pathologic convergence between TBI and AD. Increased ac-tau in human AD brain is further augmented in AD patients with history of TBI, and patients receiving the p300/CBP inhibitors salsalate or diflunisal exhibit decreased incidence of AD and clinically diagnosed TBI.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/prevention & control , Brain Injuries, Traumatic/complications , Neuroprotection , tau Proteins/metabolism , Acetylation , Alzheimer Disease/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Brain Injuries, Traumatic/metabolism , Cell Line , Diflunisal/therapeutic use , Female , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Salicylates/therapeutic use , Sirtuin 1/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism , tau Proteins/blood
2.
Neurotrauma Rep ; 2(1): 1-13, 2021.
Article in English | MEDLINE | ID: mdl-33748810

ABSTRACT

The jet-flow overpressure chamber (OPC) has been previously reported as a model of blast-mediated traumatic brain injury (bTBI). However, rigorous characterization of the features of this injury apparatus shows that it fails to recapitulate exposure to an isolated blast wave. Through combined experimental and computational modeling analysis of gas-dynamic flow conditions, we show here that the jet-flow OPC produces a collimated high-speed jet flow with extreme dynamic pressure that delivers a severe compressive impulse. Variable rupture dynamics of the diaphragm through which the jet flow originates also generate a weak and infrequent shock front. In addition, there is a component of acceleration-deceleration injury to the head as it is agitated in the headrest. Although not a faithful model of free-field blast exposure, the jet-flow OPC produces a complex multi-modal model of TBI that can be useful in laboratory investigation of putative TBI therapies and fundamental neurophysiological processes after brain injury.

3.
Antioxid Redox Signal ; 35(7): 511-530, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33501899

ABSTRACT

Aims: Impaired embryonic cortical interneuron development from prenatal stress is linked to adult neuropsychiatric impairment, stemming in part from excessive generation of reactive oxygen species in the developing embryo. Unfortunately, there are no preventive medicines that mitigate the risk of prenatal stress to the embryo, as the underlying pathophysiologic mechanisms are poorly understood. Our goal was to interrogate the molecular basis of prenatal stress-mediated damage to the embryonic brain to identify a neuroprotective strategy. Results: Chronic prenatal stress in mice dysregulated nicotinamide adenine dinucleotide (NAD+) synthesis enzymes and cortical interneuron development in the embryonic brain, leading to axonal degeneration in the hippocampus, cognitive deficits, and depression-like behavior in adulthood. Offspring were protected from these deleterious effects by concurrent maternal administration of the NAD+-modulating agent P7C3-A20, which crossed the placenta to access the embryonic brain. Prenatal stress also produced axonal degeneration in the adult corpus callosum, which was not prevented by maternal P7C3-A20. Innovation: Prenatal stress dysregulates gene expression of NAD+-synthesis machinery and GABAergic interneuron development in the embryonic brain, which is associated with adult cognitive impairment and depression-like behavior. We establish a maternally directed treatment that protects offspring from these effects of prenatal stress. Conclusion: NAD+-synthesis machinery and GABAergic interneuron development are critical to proper embryonic brain development underlying postnatal neuropsychiatric functioning, and these systems are highly susceptible to prenatal stress. Pharmacologic stabilization of NAD+ in the stressed embryonic brain may provide a neuroprotective strategy that preserves normal embryonic development and protects offspring from neuropsychiatric impairment. Antioxid. Redox Signal. 35, 511-530.


Subject(s)
Cognitive Dysfunction , Neuroprotective Agents , Prenatal Exposure Delayed Effects , Animals , Carbazoles/pharmacology , Carbazoles/therapeutic use , Female , Hippocampus , Mice , Neurogenesis , Neuroprotective Agents/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy , Stress, Psychological/complications
4.
Mol Psychiatry ; 26(6): 2286-2298, 2021 06.
Article in English | MEDLINE | ID: mdl-32332995

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by persistent fear memory of remote traumatic events, mental re-experiencing of the trauma, long-term cognitive deficits, and PTSD-associated hippocampal dysfunction. Extinction-based therapeutic approaches acutely reduce fear. However, many patients eventually relapse to the original conditioned fear response. Thus, understanding the underlying molecular mechanisms of this condition is critical to developing new treatments for patients. Mutations in the neuropsychiatric risk gene CACNA1C, which encodes the Cav1.2 isoform of the L-type calcium channel, have been implicated in both PTSD and highly comorbid neuropsychiatric conditions, such as anxiety and depression. Here, we report that male mice with global heterozygous loss of cacna1c exhibit exacerbated contextual fear that persists at remote time points (up to 180 days after shock), despite successful acute extinction training, reminiscent of PTSD patients. Because dopamine has been implicated in contextual fear memory, and Cav1.2 is a downstream target of dopamine D1-receptor (D1R) signaling, we next generated mice with specific deletion of cacna1c from D1R-expressing neurons (D1-cacna1cKO mice). Notably, D1-cacna1cKO mice also show the same exaggerated remote contextual fear, as well as persistently elevated anxiety-like behavior and impaired spatial memory at remote time points, reminiscent of chronic anxiety in treatment-resistant PTSD. We also show that D1-cacna1cKO mice exhibit elevated death of young hippocampal neurons, and that treatment with the neuroprotective agent P7C3-A20 eradicates persistent remote fear. Augmenting survival of young hippocampal neurons may thus provide an effective therapeutic approach for promoting durable remission of PTSD, particularly in patients with CACNA1C mutations or other genetic aberrations that impair calcium signaling or disrupt the survival of young hippocampal neurons.


Subject(s)
Stress Disorders, Post-Traumatic , Animals , Calcium Channels, L-Type/genetics , Conditioning, Classical , Dopamine , Extinction, Psychological , Fear , Humans , Male , Mice , Neurons , Stress Disorders, Post-Traumatic/genetics
5.
Pain ; 162(4): 1163-1175, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33027220

ABSTRACT

ABSTRACT: Chronic complications of traumatic brain injury represent one of the greatest financial burdens and sources of suffering in the society today. A substantial number of these patients suffer from posttraumatic headache (PTH), which is typically associated with tactile allodynia. Unfortunately, this phenomenon has been understudied, in large part because of the lack of well-characterized laboratory animal models. We have addressed this gap in the field by characterizing the tactile sensory profile of 2 nonpenetrating models of PTH. We show that multimodal traumatic brain injury, administered by a jet-flow overpressure chamber that delivers a severe compressive impulse accompanied by a variable shock front and acceleration-deceleration insult, produces long-term tactile hypersensitivity and widespread sensitization. These are phenotypes reminiscent of PTH in patients, in both cephalic and extracephalic regions. By contrast, closed head injury induces only transient cephalic tactile hypersensitivity, with no extracephalic consequences. Both models show a more severe phenotype with repetitive daily injury for 3 days, compared with either 1 or 3 successive injuries in a single day, providing new insight into patterns of injury that may place patients at a greater risk of developing PTH. After recovery from transient cephalic tactile hypersensitivity, mice subjected to closed head injury demonstrate persistent hypersensitivity to established migraine triggers, including calcitonin gene-related peptide and sodium nitroprusside, a nitric oxide donor. Our results offer the field new tools for studying PTH and preclinical support for a pathophysiologic role of calcitonin gene-related peptide in this condition.


Subject(s)
Brain Injuries, Traumatic , Migraine Disorders , Post-Traumatic Headache , Animals , Brain Injuries, Traumatic/complications , Calcitonin Gene-Related Peptide , Humans , Hyperalgesia/etiology , Mice , Migraine Disorders/etiology
6.
Proc Natl Acad Sci U S A ; 117(44): 27667-27675, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33087571

ABSTRACT

Chronic neurodegeneration in survivors of traumatic brain injury (TBI) is a major cause of morbidity, with no effective therapies to mitigate this progressive and debilitating form of nerve cell death. Here, we report that pharmacologic restoration of the blood-brain barrier (BBB), 12 mo after murine TBI, is associated with arrested axonal neurodegeneration and cognitive recovery, benefits that persisted for months after treatment cessation. Recovery was achieved by 30 d of once-daily administration of P7C3-A20, a compound that stabilizes cellular energy levels. Four months after P7C3-A20, electron microscopy revealed full repair of TBI-induced breaks in cortical and hippocampal BBB endothelium. Immunohistochemical staining identified additional benefits of P7C3-A20, including restoration of normal BBB endothelium length, increased brain capillary pericyte density, increased expression of BBB tight junction proteins, reduced brain infiltration of immunoglobulin, and attenuated neuroinflammation. These changes were accompanied by cessation of TBI-induced chronic axonal degeneration. Specificity for P7C3-A20 action on the endothelium was confirmed by protection of cultured human brain microvascular endothelial cells from hydrogen peroxide-induced cell death, as well as preservation of BBB integrity in mice after exposure to toxic levels of lipopolysaccharide. P7C3-A20 also protected mice from BBB degradation after acute TBI. Collectively, our results provide insights into the pathophysiologic mechanisms behind chronic neurodegeneration after TBI, along with a putative treatment strategy. Because TBI increases the risks of other forms of neurodegeneration involving BBB deterioration (e.g., Alzheimer's disease, Parkinson's disease, vascular dementia, chronic traumatic encephalopathy), P7C3-A20 may have widespread clinical utility in the setting of neurodegenerative conditions.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Injuries, Traumatic/drug therapy , Carbazoles/pharmacology , Cognition/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Blood-Brain Barrier/cytology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Carbazoles/therapeutic use , Cells, Cultured , Chronic Disease/drug therapy , Cognition/physiology , Disease Models, Animal , Endothelial Cells , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Male , Mice , Microscopy, Electron , Microvessels/cytology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Survivors
7.
ACS Chem Neurosci ; 10(3): 1595-1602, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30421909

ABSTRACT

Compounds targeting the sigma 2 receptor, which we recently cloned and showed to be identical with transmembrane protein 97 (σ2R/TMEM97), are broadly applicable therapeutic agents currently in clinical trials for imaging in breast cancer and for treatment of Alzheimer's disease and schizophrenia. These promising applications coupled with our previous observation that the σ2R/TMEM97 modulator SAS-0132 has neuroprotective attributes and improves cognition in wild-type mice suggests that modulating σ2R/TMEM97 may also have therapeutic benefits in other neurodegenerative conditions such as traumatic brain injury (TBI). Herein, we report that DKR-1677, a novel derivative of SAS-0132 with increased affinity and selectivity for σ2R/Tmem97 ( Ki = 5.1 nM), is neuroprotective after blast-induced and controlled cortical impact (CCI) TBI in mice. Specifically, we discovered that treatment with DKR-1677 decreases axonal degeneration after blast-induced TBI and enhances survival of cortical neurons and oligodendrocytes after CCI injury. Furthermore, treatment with DKR-1677 preserves cognition in the Morris water maze after blast TBI. Our results support an increasingly broad role for σ2R/Tmem97 modulation in neuroprotection and suggest a new approach for treating patients suffering from TBI.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Membrane Proteins/drug effects , Neuroprotective Agents/pharmacology , Receptors, sigma/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cognition/drug effects , Disease Models, Animal , Neurons/drug effects
8.
Transl Psychiatry ; 8(1): 202, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30258178

ABSTRACT

There is a critical need for translating basic science discoveries into new therapeutics for patients suffering from difficult to treat neuropsychiatric and neurodegenerative conditions. Previously, a target-agnostic in vivo screen in mice identified P7C3 aminopropyl carbazole as capable of enhancing the net magnitude of postnatal neurogenesis by protecting young neurons from death. Subsequently, neuroprotective efficacy of P7C3 compounds in a broad spectrum of preclinical rodent models has also been observed. An important next step in translating this work to patients is to determine whether P7C3 compounds exhibit similar efficacy in primates. Adult male rhesus monkeys received daily oral P7C3-A20 or vehicle for 38 weeks. During weeks 2-11, monkeys received weekly injection of 5'-bromo-2-deoxyuridine (BrdU) to label newborn cells, the majority of which would normally die over the following 27 weeks. BrdU+ cells were quantified using unbiased stereology. Separately in mice, the proneurogenic efficacy of P7C3-A20 was compared to that of NSI-189, a proneurogenic drug currently in clinical trials for patients with major depression. Orally-administered P7C3-A20 provided sustained plasma exposure, was well-tolerated, and elevated the survival of hippocampal BrdU+ cells in nonhuman primates without adverse central or peripheral tissue effects. In mice, NSI-189 was shown to be pro-proliferative, and P7C3-A20 elevated the net magnitude of hippocampal neurogenesis to a greater degree than NSI-189 through its distinct mechanism of promoting neuronal survival. This pilot study provides evidence that P7C3-A20 safely protects neurons in nonhuman primates, suggesting that the neuroprotective efficacy of P7C3 compounds is likely to translate to humans as well.


Subject(s)
Carbazoles/administration & dosage , Hippocampus/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Administration, Oral , Animals , Carbazoles/pharmacokinetics , Hippocampus/physiology , Macaca mulatta , Male , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Neurons/physiology , Pilot Projects
9.
Neuroscience ; 380: 90-102, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29660444

ABSTRACT

Ca2+-binding protein 1 (CaBP1) is a Ca2+-sensing protein similar to calmodulin that potently regulates voltage-gated Ca2+ channels. Unlike calmodulin, however, CaBP1 is mainly expressed in neuronal cell-types and enriched in the hippocampus, where its function is unknown. Here, we investigated the role of CaBP1 in hippocampal-dependent behaviors using mice lacking expression of CaBP1 (C-KO). By western blot, the largest CaBP1 splice variant, caldendrin, was detected in hippocampal lysates from wild-type (WT) but not C-KO mice. Compared to WT mice, C-KO mice exhibited mild deficits in spatial learning and memory in both the Barnes maze and in Morris water maze reversal learning. In contextual but not cued fear-conditioning assays, C-KO mice showed greater freezing responses than WT mice. In addition, the number of adult-born neurons in the hippocampus of C-KO mice was ∼40% of that in WT mice, as measured by bromodeoxyuridine labeling. Moreover, hippocampal long-term potentiation was significantly reduced in C-KO mice. We conclude that CaBP1 is required for cellular mechanisms underlying optimal encoding of hippocampal-dependent spatial and fear-related memories.


Subject(s)
Calcium-Binding Proteins/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Memory/physiology , Spatial Learning/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
J Stroke Cerebrovasc Dis ; 27(5): 1412-1416, 2018 May.
Article in English | MEDLINE | ID: mdl-29398531

ABSTRACT

BACKGROUND: Exogenous administration of uric acid, a naturally occurring antioxidant that scavenges reactive oxygen species in vasculature, has shown protective efficacy in both rodent models of stroke and human stroke patients in Spain as an adjuvant treatment to mechanical thrombectomy. Before clinical trials can be initiated in the United States, however, confirmation of efficacy in alternative preclinical models is required in accordance with stroke therapy academic industry roundtable-RIGOR criteria. To date, preclinical efficacy has only been established in the acute setting in male rodents. METHODS: To address this need, we subjected 7- to 9-week old ovariectomized female mice to filament-induced right middle cerebral artery ischemia and reperfusion, an established preclinical model of mechanical thrombectomy. Fidelity of the procedure was monitored by laser Doppler flowmetry. A separate lab randomly assigned animals to vehicle versus uric acid infusion, which was initiated immediately after 45 minutes of reperfusion. Poststroke analysis of infarction size and neurologic function were conducted by investigators blind to treatment group, with a 7-day primary endpoint and a 3-day intermediary analysis at 1and. RESULTS: Infarct size and neurologic function at 7 days poststroke were significantly improved in uric acid-treated animals, relative to vehicle. CONCLUSION: Efficacy of uric acid in preclinical models of stroke is now expanded to include female mice analyzed at a later time point than has been investigated previously. These results support stroke therapy academic industry roundtable-RIGOR driven determination of the suitability of acute administration of uric acid as an adjuvant to mechanical thrombectomy in clinical trials for patients with stroke.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Uric Acid/pharmacology , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Mice, Inbred C57BL , Ovariectomy , Recovery of Function , Time Factors
11.
Biol Psychiatry ; 84(7): 488-498, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29246437

ABSTRACT

BACKGROUND: In addition to cognitive deficits, Alzheimer's disease (AD) is associated with other neuropsychiatric symptoms, including severe depression. Indeed, depression often precedes cognitive deficits in patients with AD. Unfortunately, the field has seen only minimal therapeutic advances, underscoring the critical need for new treatments. P7C3 aminopropyl carbazoles promote neuronal survival by enhancing nicotinamide adenine dinucleotide flux in injured neurons. Neuroprotection with P7C3 compounds has been demonstrated in preclinical models of neurodegeneration by virtue of promoting neuronal survival independently of early disease-specific pathology, resulting in protection from cognitive deficits and depressive-like behavior. We hypothesize that P7C3 compounds might be uniquely applicable to patients with AD, given the comorbid presentation of depression and cognitive deficits. METHODS: Aging male and female wild-type and TgF344-AD rats, a well-characterized preclinical AD model, were administered (-)-P7C3-S243 daily for 9 and 18 months, beginning at 6 months of age. Behavioral phenotypes related to cognition and depression were assessed at 15 and 24 months, and brain pathology and biochemistry were assessed at 24 months. RESULTS: (-)-P7C3-S243 safely protected aging male and female wild-type and TgF344-AD rats from cognitive deficits and depressive-like behavior. Depressive-like behavior occurred earlier than cognitive deficits in TgF344-AD rats, consistent with AD in many patients. Treatment with (-)-P7C3-S243 blocked neurodegeneration in TgF344-AD rats, without altering amyloid deposition or indicators of neuroinflammation. CONCLUSIONS: Neuronal cell death-specific treatment approaches, such as P7C3 compounds, may represent a new treatment approach for patients experiencing the combination of cognitive deficits and depression associated with AD.


Subject(s)
Alzheimer Disease/drug therapy , Carbazoles/pharmacology , Cognitive Dysfunction/drug therapy , Depression/drug therapy , Neuroprotective Agents/pharmacology , Aging/drug effects , Alzheimer Disease/complications , Amyloid beta-Peptides/drug effects , Animals , Behavior, Animal/drug effects , Carbazoles/administration & dosage , Cognitive Dysfunction/etiology , Depression/etiology , Disease Models, Animal , Female , Inflammation/drug therapy , Male , Nerve Degeneration/drug therapy , Neuroglia/drug effects , Neuroprotective Agents/administration & dosage , Rats , Rats, Inbred F344 , Rats, Transgenic
12.
eNeuro ; 3(5)2016.
Article in English | MEDLINE | ID: mdl-27822499

ABSTRACT

Axonal degeneration is a prominent feature of many forms of neurodegeneration, and also an early event in blast-mediated traumatic brain injury (TBI), the signature injury of soldiers in Iraq and Afghanistan. It is not known, however, whether this axonal degeneration is what drives development of subsequent neurologic deficits after the injury. The Wallerian degeneration slow strain (WldS) of mice is resistant to some forms of axonal degeneration because of a triplicated fusion gene encoding the first 70 amino acids of Ufd2a, a ubiquitin-chain assembly factor, that is linked to the complete coding sequence of nicotinamide mononucleotide adenylyltransferase 1 (NMAT1). Here, we demonstrate that resistance of WldS mice to axonal degeneration after blast-mediated TBI is associated with preserved function in hippocampal-dependent spatial memory, cerebellar-dependent motor balance, and retinal and optic nerve-dependent visual function. Thus, early axonal degeneration is likely a critical driver of subsequent neurobehavioral complications of blast-mediated TBI. Future therapeutic strategies targeted specifically at mitigating axonal degeneration may provide a uniquely beneficial approach to treating patients suffering from the effects of blast-mediated TBI.


Subject(s)
Blast Injuries/pathology , Blast Injuries/physiopathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Wallerian Degeneration/pathology , Wallerian Degeneration/physiopathology , Animals , Axons/pathology , Axons/physiology , Blast Injuries/complications , Blast Injuries/psychology , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/psychology , Cognition , Disease Models, Animal , Male , Maze Learning , Mice, Mutant Strains , Motor Activity , Neuroprotection , Retina/pathology , Retina/physiopathology , Spatial Memory , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vision Disorders/etiology , Vision Disorders/pathology , Vision Disorders/physiopathology , Vision Disorders/psychology , Visual Perception , Wallerian Degeneration/etiology , Wallerian Degeneration/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...