Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.578
Filter
1.
PLoS One ; 19(5): e0303150, 2024.
Article in English | MEDLINE | ID: mdl-38728304

ABSTRACT

The Ang-(1-7)/MasR axis is critically involved in treating several diseases; For example, Ang-(1-7) improves inflammatory response and neurological function after traumatic brain injury and inhibits post-inflammatory hypothermia. However, its function in traumatic brain injury (TBI) combined with seawater immersion hypothermia remains unclear. Here, we used a mice model of hypothermic TBI and a BV2 cell model of hypothermic inflammation to investigate whether the Ang-(1-7)/MasR axis is involved in ameliorating hypothermic TBI. Quantitative reverse transcription PCR, western blotting assay, and immunofluorescence assay were performed to confirm microglia polarization and cytokine regulation. Hematoxylin-eosin staining, Nissl staining, and immunohistochemical assay were conducted to assess the extent of hypothermic TBI-induced damage and the ameliorative effect of Ang-(1-7) in mice. An open field experiment and neurological function scoring with two approaches were used to assess the degree of recovery and prognosis in mice. After hypothermic TBI establishment in BV2 cells, the Ang-(1-7)/MasR axis induced phenotypic transformation of microglia from M1 to M2, inhibited IL-6 and IL-1ß release, and upregulated IL-4 and IL-10 levels. After hypothermic TBI development in mice, intraperitoneally administered Ang-(1-7) attenuated histological damage and promoted neurological recovery. These findings suggest that hypothermia exacerbates TBI-induced damage and that the Ang-(1-7)/MasR axis can ameliorate hypothermic TBI and directly affect prognosis.


Subject(s)
Angiotensin I , Brain Injuries, Traumatic , Microglia , Neuroinflammatory Diseases , Peptide Fragments , Animals , Microglia/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Mice , Male , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism , Phenotype , Disease Models, Animal , Hypothermia, Induced , Cytokines/metabolism , Cell Line , Hypothermia/metabolism , Inflammation/pathology , Inflammation/metabolism
2.
J Neurosci Res ; 102(5): e25338, 2024 May.
Article in English | MEDLINE | ID: mdl-38706427

ABSTRACT

The enzyme indoleamine 2,3 dioxygenase 1 (IDO1) catalyzes the rate-limiting step in the kynurenine pathway (KP) which produces both neuroprotective and neurotoxic metabolites. Neuroinflammatory signals produced as a result of pathological conditions can increase production of IDO1 and boost its enzymatic capacity. IDO1 and the KP have been implicated in behavioral recovery after human traumatic brain injury (TBI), but their roles in experimental models of TBI are for the most part unknown. We hypothesized there is an increase in KP activity in the fluid percussion injury (FPI) model of TBI, and that administration of an IDO1 inhibitor will improve neurological recovery. In this study, adult male Sprague Dawley rats were subjected to FPI or sham injury and received twice-daily oral administration of the IDO1 inhibitor PF-06840003 (100 mg/kg) or vehicle control. FPI resulted in a significant increase in KP activity, as demonstrated by an increased ratio of kynurenine: tryptophan, in the perilesional neocortex and ipsilateral hippocampus 3 days postinjury (DPI), which normalized by 7 DPI. The increase in KP activity was prevented by PF-06840003. IDO1 inhibition also improved memory performance as assessed in the Barnes maze and anxiety behaviors as assessed in open field testing in the first 28 DPI. These results suggest increased KP activity after FPI may mediate neurological dysfunction, and IDO1 inhibition should be further investigated as a potential therapeutic target to improve recovery.


Subject(s)
Brain Injuries, Traumatic , Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenine , Rats, Sprague-Dawley , Animals , Male , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Rats , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Kynurenine/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Disease Models, Animal , Recovery of Function/drug effects , Tryptophan/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Maze Learning/drug effects
3.
Aging (Albany NY) ; 16(9): 7946-7960, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38713160

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and emerging evidence indicates that trigeminal nerve electrical stimulation (TNS) is a promising therapeutic intervention for neurological impairment following TBI. However, the precise mechanisms underlying the neuroprotective effects of TNS in TBI are poorly understood. Thus, the objective of this study was to investigate the potential involvement of the orexin-A (OX-A)/orexin receptor 1 (OX1R) mediated TLR4/NF-κB/NLRP3 signaling pathway in the neuroprotective effects of TNS in rats with TBI. METHODS: Sprague-Dawley rats were randomly assigned to four groups: sham, TBI, TBI+TNS+SB334867, and TBI+TNS. TBI was induced using a modified Feeney's method, and subsequent behavioral assessments were conducted to evaluate neurological function. The trigeminal nerve trunk was isolated, and TNS was administered following the establishment of the TBI model. The levels of neuroinflammation, brain tissue damage, and proteins associated with the OX1R/TLR4/NF-κB/NLRP3 signaling pathway were assessed using hematoxylin-eosin staining, Nissl staining, western blot analysis, quantitative real-time polymerase chain reaction, and immunofluorescence techniques. RESULTS: The findings of our study indicate that TNS effectively mitigated tissue damage, reduced brain edema, and alleviated neurological deficits in rats with TBI. Furthermore, TNS demonstrated the ability to attenuate neuroinflammation levels and inhibit the expression of proteins associated with the TLR4/NF-κB/NLRP3 signaling pathway. However, it is important to note that the aforementioned effects of TNS were reversible upon intracerebroventricular injection of an OX1R antagonist. CONCLUSION: TNS may prevent brain damage and relieve neurological deficits after a TBI by inhibiting inflammation, possibly via the TLR4/NF-κB/NLRP3 signaling pathway mediated by OX-A/OX1R.


Subject(s)
Brain Injuries, Traumatic , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Orexin Receptors , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 4 , Trigeminal Nerve , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Orexin Receptors/metabolism , Orexin Receptors/genetics , Rats , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Trigeminal Nerve/metabolism , Orexins/metabolism , Electric Stimulation Therapy/methods , Disease Models, Animal
4.
J Neuroinflammation ; 21(1): 124, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730498

ABSTRACT

Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.


Subject(s)
Anti-Inflammatory Agents , Brain Injuries, Traumatic , Brain-Gut Axis , Humans , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Brain-Gut Axis/physiology , Brain-Gut Axis/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology
5.
Mol Med Rep ; 29(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38639190

ABSTRACT

Traumatic Brain Injury (TBI) represents a significant public health challenge. Recovery from brain injury necessitates the collaborative efforts of various resident neural cells, predominantly microglia. The present study analyzed rat and mouse RNA expression micro­arrays, high­throughput RNA sequencing and single­cell sequencing data sourced from public databases. To construct an inflammation regulation network around TYRO protein tyrosine kinase­binding protein (TYROBP), to evaluate the role of TYROBP in cell death after TBI. These findings indicate that following TBI, neurons predominantly communicate with one another through the CXC chemokine ligand (CXCL) and CC chemokine ligand (CCL) signaling pathways, employing a paracrine mechanism to activate microglia. These activated microglia intensify the pathological progression of brain injury by releasing factors such as tumor necrosis factor α (TNF­α), vascular endothelial growth factor and transforming growth factor ß via the NF­κB pathway. Cells co­culture experiments demonstrated that neurons, impaired by mechanical injury, interact with microglia through non­contact mechanisms. Activated microglia secrete cytokines, including TNF­α, CXCL­8 and CCL2, which trigger an inflammatory response and facilitate neuronal apoptosis. TYROBP gene knockout in microglia was demonstrated to reduce this interaction and reduce neuronal cell apoptosis rates.


Subject(s)
Adaptor Proteins, Signal Transducing , Brain Injuries, Traumatic , Microglia , Animals , Mice , Rats , Apoptosis , Brain Injuries, Traumatic/metabolism , Inflammation/metabolism , Ligands , Mice, Inbred C57BL , Microglia/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing/metabolism
6.
Discov Med ; 36(183): 842-852, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38665032

ABSTRACT

BACKGROUND: Following traumatic brain injury (TBI), an imbalance arises in the central nervous system within the hippocampus region, resulting in the proliferation of mossy cell fibers, causing abnormal membrane discharge. Moreover, disruptions in cellular neurotransmitter secretion induce post-traumatic epilepsy. Extensive experimental and clinical data indicate that the orexin system plays a regulatory role in the hippocampal central nervous system, but the specific regulatory effects are unclear. Therefore, further experimental evaluation of its relevance is needed. OBJECTIVE: This study aims to investigate the effects of orexin receptor agonists (OXA) on the seizure threshold and intensity in controlled cortical impact (CCI) mice, and to understand the role of the orexin system in post-traumatic epilepsy (PTE). METHODS: Male C57BL/6 mice weighing 18-22 g were randomly divided into three groups: Sham, CCI, and CCI+OXA. The three groups of mice were sequentially constructed with models, implanted with electrodes, and established drug-delivery cannulas. After a 30-day recovery, the Sham and CCI groups were injected with physiological saline through the administration cannulas, while the CCI+OXA group was injected with OXA. Subsequently, all mice underwent electrical stimulation every 30 minutes for a total of 15 times. Epileptic susceptibility, duration, intensity, and cognitive changes were observed. Concurrently, the expression levels and changes of GABAergic neurons in the hippocampus of each group were examined by immunofluorescence. RESULTS: Injecting OXA into hippocampal CA1 reduces the threshold of post-traumatic seizures, prolongs the post-discharge duration, prolongs seizure duration, reduces cognitive ability, and exacerbates the loss of GABAergic neurons in the hippocampal region. CONCLUSIONS: Based on the results, we can find that injecting OXA antagonists into the CA1 region of the hippocampus can treat or prevent the occurrence and progression of post-traumatic epilepsy.


Subject(s)
Brain Injuries, Traumatic , Mice, Inbred C57BL , Orexins , Animals , Male , Mice , Orexins/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Orexin Receptors/metabolism , Epilepsy, Post-Traumatic/etiology , Epilepsy, Post-Traumatic/metabolism , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Epilepsy/etiology , Epilepsy/metabolism , Seizures/etiology , Seizures/metabolism
7.
J Neuroinflammation ; 21(1): 113, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685031

ABSTRACT

Obesity increases the morbidity and mortality of traumatic brain injury (TBI). Detailed analyses of transcriptomic changes in the brain and adipose tissue were performed to elucidate the interactive effects between high-fat diet-induced obesity (DIO) and TBI. Adult male mice were fed a high-fat diet (HFD) for 12 weeks prior to experimental TBI and continuing after injury. High-throughput transcriptomic analysis using Nanostring panels of the total visceral adipose tissue (VAT) and cellular components in the brain, followed by unsupervised clustering, principal component analysis, and IPA pathway analysis were used to determine shifts in gene expression patterns and molecular pathway activity. Cellular populations in the cortex and hippocampus, as well as in VAT, during the chronic phase after combined TBI-HFD showed amplification of central and peripheral microglia/macrophage responses, including superadditive changes in selected gene expression signatures and pathways. Furthermore, combined TBI and HFD caused additive dysfunction in Y-Maze, Novel Object Recognition (NOR), and Morris water maze (MWM) cognitive function tests. These novel data suggest that HFD-induced obesity and TBI can independently prime and support the development of altered states in brain microglia and VAT, including the disease-associated microglia/macrophage (DAM) phenotype observed in neurodegenerative disorders. The interaction between HFD and TBI promotes a shift toward chronic reactive microglia/macrophage transcriptomic signatures and associated pro-inflammatory disease-altered states that may, in part, underlie the exacerbation of cognitive deficits. Thus, targeting of HFD-induced reactive cellular phenotypes, including in peripheral adipose tissue immune cell populations, may serve to reduce microglial maladaptive states after TBI, attenuating post-traumatic neurodegeneration and neurological dysfunction.


Subject(s)
Brain Injuries, Traumatic , Brain , Cognitive Dysfunction , Diet, High-Fat , Macrophages , Mice, Inbred C57BL , Microglia , Animals , Diet, High-Fat/adverse effects , Microglia/metabolism , Microglia/pathology , Male , Mice , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Cognitive Dysfunction/metabolism , Macrophages/metabolism , Macrophages/pathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Brain/pathology , Brain/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Recognition, Psychology/physiology , Obesity/pathology , Obesity/complications , Maze Learning/physiology
8.
Brain Res ; 1834: 148907, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38570153

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI), as a major public health problem, is characterized by high incidence rate, disability rate, and mortality rate. Neuroinflammation plays a crucial role in the pathogenesis of TBI. Triggering receptor expressed on myeloid cells-1 (TREM-1) is recognized as an amplifier of the inflammation in diseases of the central nervous system (CNS). However, the function of TREM-1 remains unclear post-TBI. This study aimed to investigate the function of TREM-1 in neuroinflammation induced by TBI. METHODS: Brain water content (BWC), modified neurological severity score (mNSS), and Morris Water Maze (MWM) were measured to evaluate the effect of TREM-1 inhibition on nervous system function and outcome after TBI. TREM-1 expression in vivo was evaluated by Western blotting. The cellular localization of TREM-1 in the damaged region was observed via immunofluorescence staining. We also conducted Western blotting to examine expression of SYK, p-SYK and other downstream proteins. RESULTS: We found that inhibition of TREM-1 reduced brain edema, decreased mNSS and improved neurobehavioral outcomes after TBI. It was further determined that TREM-1 was expressed on microglia and modulated subtype transition of microglia. Inhibition of TREM-1 alleviated neuroinflammation, which was associated with SYK/p38MAPK signaling pathway. CONCLUSIONS: These findings suggest that TREM-1 can be a potential clinical therapeutic target for alleviating neuroinflammation after TBI.


Subject(s)
Brain Injuries, Traumatic , Microglia , Neuroinflammatory Diseases , Syk Kinase , Triggering Receptor Expressed on Myeloid Cells-1 , p38 Mitogen-Activated Protein Kinases , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/drug therapy , Animals , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Microglia/metabolism , Microglia/drug effects , Syk Kinase/metabolism , Syk Kinase/antagonists & inhibitors , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , p38 Mitogen-Activated Protein Kinases/metabolism , Mice , Signal Transduction/drug effects , Brain Edema/metabolism , Brain Edema/drug therapy , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL
9.
Acta Physiol (Oxf) ; 240(6): e14142, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38584589

ABSTRACT

AIM: Astrocytes respond to stressors by acquiring a reactive state characterized by changes in their morphology and function. Molecules underlying reactive astrogliosis, however, remain largely unknown. Given that several studies observed increase in the Amyloid Precursor Protein (APP) in reactive astrocytes, we here test whether APP plays a role in reactive astrogliosis. METHODS: We investigated whether APP instigates reactive astroglios by examining in vitro and in vivo the morphology and function of naive and APP-deficient astrocytes in response to APP and well-established stressors. RESULTS: Overexpression of APP in cultured astrocytes led to remodeling of the intermediate filament network, enhancement of cytokine production, and activation of cellular programs centered around the interferon (IFN) pathway, all signs of reactive astrogliosis. Conversely, APP deletion abrogated remodeling of the intermediate filament network and blunted expression of IFN-stimulated gene products in response to lipopolysaccharide. Following traumatic brain injury (TBI), mouse reactive astrocytes also exhibited an association between APP and IFN, while APP deletion curbed the increase in glial fibrillary acidic protein observed canonically in astrocytes in response to TBI. CONCLUSIONS: The APP thus represents a candidate molecular inducer and regulator of reactive astrogliosis. This finding has implications for understanding pathophysiology of neurodegenerative and other diseases of the nervous system characterized by reactive astrogliosis and opens potential new therapeutic avenues targeting APP and its pathways to modulate reactive astrogliosis.


Subject(s)
Amyloid beta-Protein Precursor , Astrocytes , Gliosis , Animals , Gliosis/metabolism , Gliosis/pathology , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Astrocytes/metabolism , Astrocytes/pathology , Mice , Cells, Cultured , Mice, Inbred C57BL , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Mice, Knockout
10.
Brain Res ; 1835: 148908, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38582416

ABSTRACT

BDNF, a neurotrophic factor, and its receptors have been implicated in the pathophysiology of mild traumatic brain injury (mTBI). The brainstem houses many vital functions, that are also associated with signs and symptoms of mTBI, but has been understudied in mTBI animal models. We determined the extent to which neurotrophic protein and associated receptor expression is affected within the brainstem of adult rats following mTBI. Their behavioral function was assessed and temporal expression of the 'negative' regulators of neuronal function (p75, t-TrkB, and pro-BDNF) and 'positive' neuroprotective (FL-TrkB and m-BDNF) protein isoforms were determined via western blot and immunohistochemistry at 1, 3, 7, and 14 post-injury days (PID) following mTBI or sham (control) procedure. Within the brainstem, p75 expression increased at PID 1 vs. sham animals. t-TrkB and pro-BDNF expression increased at PID 7 and 14. The 'positive' protein isoforms of FL-TrkB and m-BDNF expression were increased only at PID 7. The ratio of t-TrkB:FL-TrkB (negative:positive) was substantial across groups and time points, suggesting a negative impact of neurotrophic signaling on neuronal function. Additional NeuN experiments revealed cell death occurring within a subset of neurons within the medulla. While behavioral measures improved by PID 7-14, negative neurotrophic biochemical responses persisted. Despite the assertion that mTBI produces "mild" injury, evidence of cell death was observed in the medulla. Ratios of TrkB and BDNF isoforms with conflicting functions suggest that future work should specifically measure each subtype since they induce opposing downstream effects on neuronal function.


Subject(s)
Brain Stem , Brain-Derived Neurotrophic Factor , Rats, Sprague-Dawley , Receptor, trkB , Animals , Male , Brain-Derived Neurotrophic Factor/metabolism , Receptor, trkB/metabolism , Brain Stem/metabolism , Rats , Brain Concussion/metabolism , Disease Models, Animal , Neurons/metabolism , Protein Isoforms/metabolism , Time Factors , Nerve Tissue Proteins/metabolism , Brain Injuries, Traumatic/metabolism
11.
Redox Biol ; 72: 103156, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640584

ABSTRACT

Regulation of the oxidative stress response is crucial for the management and prognosis of traumatic brain injury (TBI). The copper chaperone Antioxidant 1 (Atox1) plays a crucial role in regulating intracellular copper ion balance and impacting the antioxidant capacity of mitochondria, as well as the oxidative stress state of cells. However, it remains unknown whether Atox1 is involved in modulating oxidative stress following TBI. Here, we investigated the regulatory role of Atox1 in oxidative stress on neurons both in vivo and in vitro, and elucidated the underlying mechanism through culturing hippocampal HT-22 cells with Atox1 mutation. The expression of Atox1 was significantly diminished following TBI, while mice with overexpressed Atox1 exhibited a more preserved hippocampal structure and reduced levels of oxidative stress post-TBI. Furthermore, the mice displayed notable impairments in learning and memory functions after TBI, which were ameliorated by the overexpression of Atox1. In the stretch injury model of HT-22 cells, overexpression of Atox1 mitigated oxidative stress by preserving the normal morphology and network connectivity of mitochondria, as well as facilitating the elimination of damaged mitochondria. Mechanistically, co-immunoprecipitation and mass spectrometry revealed the binding of Atox1 to DJ-1. Knockdown of DJ-1 in HT-22 cells significantly impaired the antioxidant capacity of Atox1. Mutations in the copper-binding motif or sequestration of free copper led to a substantial decrease in the interaction between Atox1 and DJ-1, with overexpression of DJ-1 failing to restore the antioxidant capacity of Atox1 mutants. The findings suggest that DJ-1 mediates the ability of Atox1 to withstand oxidative stress. And targeting Atox1 could be a potential therapeutic approach for addressing post-traumatic neurological dysfunction.


Subject(s)
Brain Injuries, Traumatic , Copper Transport Proteins , Hippocampus , Mitophagy , Neurons , Oxidative Stress , Protein Deglycase DJ-1 , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/genetics , Mice , Hippocampus/metabolism , Hippocampus/pathology , Neurons/metabolism , Protein Deglycase DJ-1/metabolism , Protein Deglycase DJ-1/genetics , Copper Transport Proteins/metabolism , Copper Transport Proteins/genetics , Mitochondria/metabolism , Disease Models, Animal , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Male , Antioxidants/metabolism , Cell Line , Humans
12.
Cell Mol Neurobiol ; 44(1): 41, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38656449

ABSTRACT

The cadherin family plays a pivotal role in orchestrating synapse formation in the central nervous system. Cadherin-related family member 1 (CDHR1) is a photoreceptor-specific calmodulin belonging to the expansive cadherin superfamily. However, its role in traumatic brain injury (TBI) remains largely unknown. CDHR1 expression across various brain tissue sites was analyzed using the GSE104687 dataset. Employing a summary-data-based Mendelian Randomization (SMR) approach, integrated analyses were performed by amalgamating genome-wide association study abstracts from TBI with public data on expressed quantitative trait loci and DNA methylation QTL from both blood and diverse brain tissues. CDHR1 expression and localization in different brain tissues were meticulously delineated using western blotting, immunohistochemistry, and enzyme-linked immunosorbent assay. CDHR1 expression was consistently elevated in the TBI group compared to that in the sham group across multiple tissues. The inflammatory response emerged as a crucial biological mechanism, and pro-inflammatory and anti-inflammatory factors were not expressed in either group. Integrated SMR analyses encompassing both blood and brain tissues substantiated the heightened CDHR1 expression profiles, with methylation modifications emerging as potential contributing factors for increased TBI risk. This was corroborated by western blotting and immunohistochemistry, confirming augmented CDHR1 expression following TBI. This multi-omics-based genetic association study highlights the elevated TBI risk associated with CDHR1 expression coupled with putative methylation modifications. These findings provide compelling evidence for future targeted investigations and offer promising avenues for developing interventional therapies for TBI.


Subject(s)
Brain Injuries, Traumatic , Cadherins , Animals , Humans , Male , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Cadherin Related Proteins , Cadherins/genetics , Cadherins/metabolism , DNA Methylation/genetics , Genome-Wide Association Study , Quantitative Trait Loci/genetics
13.
Neurochem Int ; 176: 105741, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38621511

ABSTRACT

Navigating menopause involves traversing a complex terrain of hormonal changes that extend far beyond reproductive consequences. Menopausal transition is characterized by a decrease in estradiol-17ß (E2), and the impact of menopause resonates not only in the reproductive system but also through the central nervous system, musculoskeletal, and gastrointestinal domains. As women undergo menopausal transition, they become more susceptible to frailty, amplifying the risk and severity of injuries, including traumatic brain injury (TBI). Menopause triggers a cascade of changes leading to a decline in muscle mass, accompanied by diminished tone and excitability, thereby restricting the availability of irisin, a crucial hormone derived from muscles. Concurrently, bone mass undergoes reduction, culminating in the onset of osteoporosis and altering the dynamics of osteocalcin, a hormone originating from bones. The diminishing levels of E2 during menopause extend their influence on the gut microbiota, resulting in a reduction in the availability of tyrosine, tryptophan, and serotonin metabolites, affecting neurotransmitter synthesis and function. Understanding the interplay between menopause, frailty, E2 decline, and the intricate metabolisms of bone, gut, and muscle is imperative when unraveling the nuances of TBI after menopause. The current review underscores the significance of accounting for menopause-associated frailty in the incidence and consequences of TBI. The review also explores potential mechanisms to enhance gut, bone, and muscle health in menopausal women, aiming to mitigate frailty and improve TBI outcomes.


Subject(s)
Brain Injuries, Traumatic , Frailty , Menopause , Humans , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Female , Menopause/metabolism , Menopause/physiology , Frailty/metabolism , Estradiol/metabolism
14.
J Mol Histol ; 55(3): 329-348, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38609527

ABSTRACT

Neural stem cell secretome (NSC-S) plays an important role in neuroprotection and recovery. Studies have shown that endoplasmic reticulum stress (ER stress) is involved in the progression of traumatic brain injury (TBI) and is a crucial cause of secondary damage and neuronal death after brain injury. Whether NSC-S is engaged in ER stress and ER stress-mediated neuronal apoptosis post-TBI has not been investigated. In the study, the Feeney SD male rat model was established. The results showed that NSC-S treatment significantly improved the behavior of rats with TBI. In addition, NSC-S relieved ER stress in TBI rats and was observed by transmission electron microscopy and western blot. The specific mechanism was further elucidated that restoration was achieved by alleviating the PERK-eIF2α pathway and thus protecting neurons from apoptosis. Notably, the discovery of calumenin (CALU) in NSC-S by liquid chromatography-tandem mass spectrometry (LC-MS/MS/MS) may be related to the protective effect of NSC-S on ER stress in neurons. Also, the mechanism by which it functions may be related to ubiquitination. In summary, NSC-S improved prognosis and ER stress in TBI rats and might be a promising treatment for relieving TBI.


Subject(s)
Apoptosis , Brain Injuries, Traumatic , Disease Models, Animal , Endoplasmic Reticulum Stress , Neural Stem Cells , Neurons , Rats, Sprague-Dawley , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Neural Stem Cells/metabolism , Rats , Humans , Neurons/metabolism , Male
15.
Sci Transl Med ; 16(743): eadk9129, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630849

ABSTRACT

Traumatic brain injury (TBI) leads to skeletal changes, including bone loss in the unfractured skeleton, and paradoxically accelerates healing of bone fractures; however, the mechanisms remain unclear. TBI is associated with a hyperadrenergic state characterized by increased norepinephrine release. Here, we identified the ß2-adrenergic receptor (ADRB2) as a mediator of skeletal changes in response to increased norepinephrine. In a murine model of femoral osteotomy combined with cortical impact brain injury, TBI was associated with ADRB2-dependent enhanced fracture healing compared with osteotomy alone. In the unfractured 12-week-old mouse skeleton, ADRB2 was required for TBI-induced decrease in bone formation and increased bone resorption. Adult 30-week-old mice had higher bone concentrations of norepinephrine, and ADRB2 expression was associated with decreased bone volume in the unfractured skeleton and better fracture healing in the injured skeleton. Norepinephrine stimulated expression of vascular endothelial growth factor A and calcitonin gene-related peptide-α (αCGRP) in periosteal cells through ADRB2, promoting formation of osteogenic type-H vessels in the fracture callus. Both ADRB2 and αCGRP were required for the beneficial effect of TBI on bone repair. Adult mice deficient in ADRB2 without TBI developed fracture nonunion despite high bone formation in uninjured bone. Blocking ADRB2 with propranolol impaired fracture healing in mice, whereas the ADRB2 agonist formoterol promoted fracture healing by regulating callus neovascularization. A retrospective cohort analysis of 72 patients with long bone fractures indicated improved callus formation in 36 patients treated with intravenous norepinephrine. These findings suggest that ADRB2 is a potential therapeutic target for promoting bone healing.


Subject(s)
Brain Injuries, Traumatic , Fractures, Bone , Humans , Animals , Mice , Fracture Healing/physiology , Vascular Endothelial Growth Factor A , Adrenergic Agents , Retrospective Studies , Brain Injuries, Traumatic/metabolism , Neovascularization, Pathologic , Norepinephrine
16.
Biomolecules ; 14(4)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38672403

ABSTRACT

Vascular contribution to cognitive impairment and dementia (VCID) is a term referring to all types of cerebrovascular and cardiovascular disease-related cognitive decline, spanning many neuroinflammatory diseases including traumatic brain injury (TBI). This becomes particularly important during mild-to-moderate TBI (m-mTBI), which is characterized by short-term memory (STM) decline. Enhanced cerebrovascular permeability for proteins is typically observed during m-mTBI. We have previously shown that an increase in the blood content of fibrinogen (Fg) during m-mTBI results in enhanced cerebrovascular permeability. Primarily extravasated via a transcellular pathway, Fg can deposit into the parenchyma and exacerbate inflammatory reactions that can lead to neurodegeneration, resulting in cognitive impairment. In the current study, we investigated the effect of a chronic reduction in Fg concentration in blood on cerebrovascular permeability and the interactions of extravasated Fg with astrocytes and neurons. Cortical contusion injury (CCI) was used to generate m-mTBI in transgenic mice with a deleted Fg γ chain (Fg γ+/-), resulting in a low blood content of Fg, and in control C57BL/6J wild-type (WT) mice. Cerebrovascular permeability was tested in vivo. Interactions of Fg with astrocytes and neurons and the expression of neuronal nuclear factor-кB (NF-кB) were assessed via immunohistochemistry. The results showed that 14 days after CCI, there was less cerebrovascular permeability, lower extravascular deposition of Fg, less activation of astrocytes, less colocalization of Fg with neurons, and lower expression of neuronal pro-inflammatory NF-кB in Fg γ+/- mice compared to that found in WT mice. Combined, our data provide strong evidence that increased Fg extravasation, and its resultant extravascular deposition, triggers astrocyte activation and leads to potential interactions of Fg with neurons, resulting in the overexpression of neuronal NF-кB. These effects suggest that reduced blood levels of Fg can be beneficial in mitigating the STM reduction seen in m-mTBI.


Subject(s)
Brain Injuries, Traumatic , Fibrinogen , Mice, Inbred C57BL , Mice, Knockout , Animals , Fibrinogen/metabolism , Fibrinogen/genetics , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/genetics , Mice , Blood-Brain Barrier/metabolism , Astrocytes/metabolism , Male , Capillary Permeability , Heterozygote , Neurons/metabolism , Disease Models, Animal
17.
Biochem Pharmacol ; 224: 116244, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685280

ABSTRACT

Traumatic brain injury (TBI) triggers a bevy of changes including mitochondrial dysfunction, apoptosis, oxidative stress, neurobehavioural impairment, and neuroinflammation, among others. Dantrolene (DNT), a muscle relaxant which inhibits intracellular Ca2+ signaling from the ER, has been repurposed as a potential neuroprotective agent in various neurological diseases. However, there have been limited studies on whether it can mitigate TBI-induced deficits and restore impaired mitochondrial dynamics. This study sought to evaluate whether Dantrolene can potentially provide neuroprotection in an in vivo model of TBI. Male wistar rats subjected to TBI were treated with DNT (10 mg/kg) 1 h and 12 h post surgery. Animals were assessed 24 h post-TBI to evaluate neurobehavioural deficits and cerebral edema. We evaluated the protein expressions of apoptotic, autophagic, and neuroinflammatory markers by immunoblotting, as well as Mitochondrial Membrane Potential (MMP) and Reactive Oxygen Species (ROS) via Flow Cytometry to ascertain the effects of DNT on TBI. We further analysed immunofluorescence staining with Glial Fibrillary Acidic Protein (GFAP) and immunohistochemistry with NF-κß to investigate neuroinflammation. H&E staining was also performed post-TBI. Our findings revealed DNT administration inhibits mitochondria-mediated apoptotis and reduces heightened oxidative stress. DNT treatment was also found to reverse neurobehavioural impairments and offer neuroprotection by preserving neuronal architechture. We also demonstrated that DNT inhibits neuronal autophagy and alleviates neuroinflammation following TBI by modulating the NF-κß/Akt signaling pathway. Thus, our results suggest a novel application of DNT in ameliorating the multitude of deficits induced by TBI, thereby conferring neuroprotection.


Subject(s)
Brain Injuries, Traumatic , Dantrolene , Mitochondria , NF-kappa B , Neuroinflammatory Diseases , Proto-Oncogene Proteins c-akt , Rats, Wistar , Animals , Dantrolene/pharmacology , Dantrolene/therapeutic use , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Male , Rats , Proto-Oncogene Proteins c-akt/metabolism , Neuroinflammatory Diseases/drug therapy , NF-kappa B/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Muscle Relaxants, Central/pharmacology , Muscle Relaxants, Central/therapeutic use
18.
J Biol Inorg Chem ; 29(2): 187-199, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38607392

ABSTRACT

Aß42 plaque formation is one of the preliminary pathologic events that occur post traumatic brain injury (TBI) which is also among the most noteworthy hallmarks of AD. Their pre symptomatic detection is therefore vital for better disease management. Chalcone-picolinic acid chelator derivative, 6-({[(6-carboxypyridin-2-yl)methyl](2-{4-[(2E)-3-[4-(dimethyl amino)phenyl]prop-2-enoyl]phenoxy}ethyl)amino}methyl)pyridine-2-carboxylic acid, Py-chal was synthesized to selectively identify amyloid plaques formed post head trauma using SPECT imaging by stable complexation to 99mTc with > 97% efficiency without compromising amyloid specificity. The binding potential of the Py-chal ligand to amyloid plaques remained high as confirmed by in vitro binding assay and photophysical spectra. Further, the Py-chal complex stained amyloid aggregates in the brain sections of rmTBI mice model. In vivo scintigraphy in TBI mice model displayed high uptake followed by high retention while the healthy rabbits displayed higher brain uptake followed by a rapid washout attributed to absence of amyloid plaques. Higher uptake in brain of TBI model was also confirmed by ex vivo biodistribution analysis wherein brain uptake of 3.38 ± 0.2% ID/g at 2 min p.i. was observed for TBI mice model. This was followed by prolonged retention and more than twofold higher activity as compared to sham mice brain. This preliminary data suggests the specificity of the radiotracer for amyloid detection post head trauma and applicability of 99mTc labeled Py-chal complex for TBI-induced ß-amyloid SPECT imaging.


Subject(s)
Amyloid beta-Peptides , Tomography, Emission-Computed, Single-Photon , Animals , Amyloid beta-Peptides/metabolism , Mice , Technetium/chemistry , Tissue Distribution , Chalcone/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/pharmacokinetics , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Craniocerebral Trauma/diagnostic imaging , Male , Brain/diagnostic imaging , Brain/metabolism
19.
Front Immunol ; 15: 1343364, 2024.
Article in English | MEDLINE | ID: mdl-38558799

ABSTRACT

Macrophage/microglia function as immune defense and homeostatic cells that originate from bone marrow progenitor cells. Macrophage/microglia activation is historically divided into proinflammatory M1 or anti-inflammatory M2 states based on intracellular dynamics and protein production. The polarization of macrophages/microglia involves a pivotal impact in modulating the development of inflammatory disorders, namely lung and traumatic brain injuries. Recent evidence indicates shared signaling pathways in lung and traumatic brain injuries, regulated through non-coding RNAs (ncRNAs) loaded into extracellular vesicles (EVs). This packaging protects ncRNAs from degradation. These vesicles are subcellular components released through a paracellular mechanism, constituting a group of nanoparticles that involve exosomes, microvesicles, and apoptotic bodies. EVs are characterized by a double-layered membrane and are abound with proteins, nucleic acids, and other bioactive compounds. ncRNAs are RNA molecules with functional roles, despite their absence of coding capacity. They actively participate in the regulation of mRNA expression and function through various mechanisms. Recent studies pointed out that selective packaging of ncRNAs into EVs plays a role in modulating distinct facets of macrophage/microglia polarization, under conditions of lung and traumatic brain injuries. This study will explore the latest findings regarding the role of EVs in the progression of lung and traumatic brain injuries, with a specific focus on the involvement of ncRNAs within these vesicles. The conclusion of this review will emphasize the clinical opportunities presented by EV-ncRNAs, underscoring their potential functions as both biomarkers and targets for therapeutic interventions.


Subject(s)
Brain Injuries, Traumatic , Extracellular Vesicles , Humans , Microglia/metabolism , Macrophages/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Brain Injuries, Traumatic/metabolism , Lung/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
20.
Cell Stem Cell ; 31(4): 519-536.e8, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38579683

ABSTRACT

Traumatic brain injury (TBI) strongly correlates with neurodegenerative disease. However, it remains unclear which neurodegenerative mechanisms are intrinsic to the brain and which strategies most potently mitigate these processes. We developed a high-intensity ultrasound platform to inflict mechanical injury to induced pluripotent stem cell (iPSC)-derived cortical organoids. Mechanically injured organoids elicit classic hallmarks of TBI, including neuronal death, tau phosphorylation, and TDP-43 nuclear egress. We found that deep-layer neurons were particularly vulnerable to injury and that TDP-43 proteinopathy promotes cell death. Injured organoids derived from C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) patients displayed exacerbated TDP-43 dysfunction. Using genome-wide CRISPR interference screening, we identified a mechanosensory channel, KCNJ2, whose inhibition potently mitigated neurodegenerative processes in vitro and in vivo, including in C9ORF72 ALS/FTD organoids. Thus, targeting KCNJ2 may reduce acute neuronal death after brain injury, and we present a scalable, genetically flexible cerebral organoid model that may enable the identification of additional modifiers of mechanical stress.


Subject(s)
Amyotrophic Lateral Sclerosis , Brain Injuries, Traumatic , Frontotemporal Dementia , Neurodegenerative Diseases , Potassium Channels, Inwardly Rectifying , Humans , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/pathology , Brain/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , C9orf72 Protein/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/etiology , Frontotemporal Dementia/pathology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...