Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.449
Filter
1.
Mol Biol Rep ; 51(1): 695, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796674

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of disability worldwide, with an estimated annual incidence of 27-69 million. TBI is a severe condition that can lead to high mortality rates and long-term cognitive, behavioral, and physical impairments in young adults. It is a significant public health concern due to the lack of effective treatments available. Quercetin, a natural flavonoid found in various fruits and vegetables, has demonstrated therapeutic potential with anti-inflammatory, antioxidant, and neuroprotective properties. Recently, some evidence has accentuated the ameliorating effects of quercetin on TBI. This review discusses quercetin's ability to reduce TBI-related damage by regulating many cellular and molecular pathways. Quercetin in vitro and in vivo studies exhibit promise in reducing inflammation, oxidative stress, apoptosis, and enhancing cognitive function post-TBI. Further clinical investigation into quercetin's therapeutic potential as a readily available adjuvant in the treatment of TBI is warranted in light of these findings. This review adds to our knowledge of quercetin's potential in treating TBI by clarifying its mechanisms of action.


Subject(s)
Antioxidants , Brain Injuries, Traumatic , Neuroprotective Agents , Oxidative Stress , Quercetin , Quercetin/pharmacology , Quercetin/therapeutic use , Brain Injuries, Traumatic/drug therapy , Humans , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Apoptosis/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
2.
J Neuroinflammation ; 21(1): 141, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807149

ABSTRACT

The lectin pathway (LP) of complement mediates inflammatory processes linked to tissue damage and loss of function following traumatic brain injury (TBI). LP activation triggers a cascade of proteolytic events initiated by LP specific enzymes called MASPs (for Mannan-binding lectin Associated Serine Proteases). Elevated serum and brain levels of MASP-2, the effector enzyme of the LP, were previously reported to be associated with the severity of tissue injury and poor outcomes in patients with TBI. To evaluate the therapeutic potential of LP inhibition in TBI, we first conducted a pilot study testing the effect of an inhibitory MASP-2 antibody (α-MASP-2), administered systemically at 4 and 24 h post-TBI in a mouse model of controlled cortical impact (CCI). Treatment with α-MASP-2 reduced sensorimotor and cognitive deficits for up to 5 weeks post-TBI. As previous studies by others postulated a critical role of MASP-1 in LP activation, we conducted an additional study that also assessed treatment with an inhibitory MASP-1 antibody (α-MASP-1). A total of 78 mice were treated intraperitoneally with either α-MASP-2, or α-MASP-1, or an isotype control antibody 4 h and 24 h after TBI or sham injury. An amelioration of the cognitive deficits assessed by Barnes Maze, prespecified as the primary study endpoint, was exclusively observed in the α-MASP-2-treated group. The behavioral data were paralleled by a reduction of the lesion size when evaluated histologically and by reduced systemic LP activity. Our data suggest that inhibition of the LP effector enzyme MASP-2 is a promising treatment strategy to limit neurological deficits and tissue loss following TBI. Our work has translational value because a MASP-2 antibody has already completed multiple late-stage clinical trials in other indications and we used a clinically relevant treatment protocol testing the therapeutic mechanism of MASP-2 inhibition in TBI.


Subject(s)
Brain Injuries, Traumatic , Disease Models, Animal , Mannose-Binding Protein-Associated Serine Proteases , Mice, Inbred C57BL , Animals , Mannose-Binding Protein-Associated Serine Proteases/antagonists & inhibitors , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/psychology , Mice , Male , Cognition Disorders/etiology , Cognition Disorders/drug therapy , Maze Learning/drug effects , Maze Learning/physiology
3.
Medicine (Baltimore) ; 103(20): e38172, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758901

ABSTRACT

This study aims to investigate the effect of amantadine use on neurological outcomes and mortality in patients with severe traumatic brain injury (TBI) (Glasgow coma score [GCS] between 3 and 8) who have been followed up on mechanical ventilators in the intensive care unit (ICU). Data from the hospital's electronic records were retrospectively searched. Patients over 18 years of age, with severe brain trauma (GCS between 3-8), who were treated with endotracheal intubation and invasive mechanical ventilation at admission to the ICU, and who were treated with Amantadine hydrochloride at least once in the first week of follow-up were included in the study. To evaluate the patients' neurological outcomes, the GCS and FOUR scores were used. GCS and FOUR scores were recorded on the 1st, 3rd, and 7th days of the first week. In addition, the score difference between the 1st and 7th day was calculated for both scores. The patients were divided into 2 groups: those receiving amantadine treatment (Group A, n = 44) and the control group (Group C, n = 47). The median age of all patients was 39 (18-81) (P = .425). When Group A and Group C were compared, no statistically significant results were found between the 1st, 3rd, and 7th day GCS values (P = .474, P = .483, and P = 329, respectively). However, the difference in GCS values between day 1 and day 7 (∆ GCS 7-1) was statistically significant (P = .012). Similarly, when Group A and Group C were compared, no statistically significant results were found between the 1st, 3rd, and 7th day FOUR score values (P = .948, P = .471, and P = .057, respectively). However, the FOUR score values between day 1 and day 7 (∆ FOUR score 7-1) were statistically significant (P = .004). There was no statistically significant difference among the groups in terms of ICU length of stay, duration of non-ICU hospital stay, and length of hospital stay (P = .222, P = .175, and P = .067, respectively). Amantadine hydrochloride may help improve neurological outcomes in patients with severe TBI. However, further research is needed to investigate this topic.


Subject(s)
Amantadine , Glasgow Coma Scale , Intensive Care Units , Respiration, Artificial , Humans , Amantadine/therapeutic use , Respiration, Artificial/statistics & numerical data , Male , Female , Middle Aged , Adult , Retrospective Studies , Intensive Care Units/statistics & numerical data , Aged , Adolescent , Aged, 80 and over , Brain Injuries, Traumatic/mortality , Brain Injuries, Traumatic/drug therapy , Young Adult , Treatment Outcome , Craniocerebral Trauma/mortality
4.
J Nanobiotechnology ; 22(1): 251, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750597

ABSTRACT

BACKGROUND: Hypothermia is a promising therapy for traumatic brain injury (TBI) in the clinic. However, the neuroprotective outcomes of hypothermia-treated TBI patients in clinical studies are inconsistent due to several severe side effects. Here, an injectable refrigerated hydrogel was designed to deliver 3-iodothyronamine (T1AM) to achieve a longer period of local hypothermia for TBI treatment. Hydrogel has four advantages: (1) It can be injected into injured sites after TBI, where it forms a hydrogel and avoids the side effects of whole-body cooling. (2) Hydrogels can biodegrade and be used for controlled drug release. (3) Released T1AM can induce hypothermia. (4) This hydrogel has increased medical value given its simple operation and ability to achieve timely treatment. METHODS: Pol/T hydrogels were prepared by a low-temperature mixing method and characterized. The effect of the Pol/T hydrogel on traumatic brain injury in mice was studied. The degradation of the hydrogel at the body level was observed with a small animal imager. Brain temperature and body temperature were measured by brain thermometer and body thermometer, respectively. The apoptosis of peripheral nerve cells was detected by immunohistochemical staining. The protective effect of the hydrogels on the blood-brain barrier (BBB) after TBI was evaluated by the Evans blue penetration test. The protective effect of hydrogel on brain edema after injury in mice was detected by Magnetic resonance (MR) in small animals. The enzyme linked immunosorbent assay (ELISA) method was used to measure the levels of inflammatory factors. The effects of behavioral tests on the learning ability and exercise ability of mice after injury were evaluated. RESULTS: This hydrogel was able to cool the brain to hypothermia for 12 h while maintaining body temperature within the normal range after TBI in mice. More importantly, hypothermia induced by this hydrogel leads to the maintenance of BBB integrity, the prevention of cell death, the reduction of the inflammatory response and brain edema, and the promotion of functional recovery after TBI in mice. This cooling method could be developed as a new approach for hypothermia treatment in TBI patients. CONCLUSION: Our study showed that injectable and biodegradable frozen Pol/T hydrogels to induce local hypothermia in TBI mice can be used for the treatment of traumatic brain injury.


Subject(s)
Blood-Brain Barrier , Brain Injuries, Traumatic , Hydrogels , Hypothermia, Induced , Animals , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/drug therapy , Mice , Hydrogels/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Male , Hypothermia, Induced/methods , Neuroprotection/drug effects , Brain/pathology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Body Temperature , Mice, Inbred C57BL
5.
Cells ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38727269

ABSTRACT

The histone deacetylase inhibitor (HDACi) valproic acid (VPA) has neuroprotective and anti-inflammatory effects in experimental traumatic brain injury (TBI), which have been partially attributed to the epigenetic disinhibition of the transcription repressor RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). Additionally, VPA changes post-traumatic brain injury (TBI) brain metabolism to create a neuroprotective environment. To address the interconnection of neuroprotection, metabolism, inflammation and REST/NRSF after TBI, we subjected C57BL/6N mice to experimental TBI and intraperitoneal VPA administration or vehicle solution at 15 min, 1, 2, and 3 days post-injury (dpi). At 7 dpi, TBI-induced an up-regulation of REST/NRSF gene expression and HDACi function of VPA on histone H3 acetylation were confirmed. Neurological deficits, brain lesion size, blood-brain barrier permeability, or astrogliosis were not affected, and REST/NRSF target genes were only marginally influenced by VPA. However, VPA attenuated structural damage in the hippocampus, microgliosis and expression of the pro-inflammatory marker genes. Analyses of plasma lipidomic and polar metabolomic patterns revealed that VPA treatment increased lysophosphatidylcholines (LPCs), which were inversely associated with interleukin 1 beta (Il1b) and tumor necrosis factor (Tnf) gene expression in the brain. The results show that VPA has mild neuroprotective and anti-inflammatory effects likely originating from favorable systemic metabolic changes resulting in increased plasma LPCs that are known to be actively taken up by the brain and function as carriers for neuroprotective polyunsaturated fatty acids.


Subject(s)
Brain Injuries, Traumatic , Inflammation , Lysophosphatidylcholines , Mice, Inbred C57BL , Neurons , Valproic Acid , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Mice , Male , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Inflammation/pathology , Inflammation/drug therapy , Lysophosphatidylcholines/blood , Cell Death/drug effects , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Repressor Proteins/metabolism , Repressor Proteins/genetics
6.
Neuroreport ; 35(9): 549-557, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38739900

ABSTRACT

Neuroinflammation after traumatic brain injury (TBI) exhibits a strong correlation with neurological impairment, which is a crucial target for improving the prognosis of TBI patients. The involvement of CXCL5/CXCR2 signaling in the regulation of neuroinflammation in brain injury models has been documented. Therefore, the effects of CXCL5 on post-TBI neuroinflammation and its potential mechanisms need to be explored. Following TBI, C57BL/6 mice were administered intraperitoneal injections of a CXCL5 neutralizing antibody (Nab-CXCL5) (5 mg/kg, 2 times/day). Subsequently, the effects on neuroinflammation, nerve injury, and neurological function were assessed. Nab-CXCL5 significantly reduced the release of inflammatory factors, inhibited the formation of inflammatory microglia and astrocytes, and reduced the infiltration of peripheral immune cells in TBI mice. Additionally, this intervention led to a reduction in neuronal impairment and facilitated the restoration of sensorimotor abilities, as well as improvements in learning and memory functions. Peripheral administration of the Nab-CXCL5 to TBI mice could suppress neuroinflammation, reduce neurological damage, and improve neurological function. Our data suggest that neutralizing antibodies against CXCL5 (Nab-CXCL5) may be a promising agent for treating TBI.


Subject(s)
Brain Injuries, Traumatic , Chemokine CXCL5 , Mice, Inbred C57BL , Neuroinflammatory Diseases , Recovery of Function , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/drug therapy , Chemokine CXCL5/metabolism , Neuroinflammatory Diseases/drug therapy , Mice , Male , Recovery of Function/drug effects , Recovery of Function/physiology , Antibodies, Neutralizing/pharmacology , Microglia/drug effects , Microglia/metabolism
7.
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
8.
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
9.
Article in Russian | MEDLINE | ID: mdl-38676685

ABSTRACT

OBJECTIVE: Evaluation of the effect of pharmacological modulation of the rehabilitation process with the drug mexidol as an adjuvant component of the rehabilitation treatment of cognitive-emotional disorders in patients who have suffered acute cerebral insufficiency (ACI) due to acute cerebrovascular accident or traumatic brain injury. MATERIAL AND METHODS: The study was conducted as a randomized interventional prospective study and consisted of 5 visits. Patients were divided into 2 groups: main (n=30, standard therapy + Mexidol IV 500 mg per day for 10 days, followed by Mexidol FORTE 250 orally, 1 tablet 3 times a day for 8 weeks) and control (n=30, standard therapy for 66 days). RESULTS: The study randomized 60 patients who underwent ACN and received rehabilitation treatment in accordance with regional routing. In the main group, there was an improvement in cognitive functions comparable to the control group (p<0.001, in both groups there was an improvement in the Schulte test «work efficiency¼ and «total execution time¼, according to the MoCA scale (visit 5 - 23.8±2.6 vs 22.9±31, p=0.227). A significant superiority of the main group over the control group was shown in such indicators as a decrease in anxiety (according to the HADS scale) (visit 4 - 2.6±2.4 vs 4.4±2.4, p=0.004), a decrease in the severity of depression (according to the Beck scale) (visit 3 - 7.5±4.5 vs 11.4±5.6, p=0.005). There was a tendency for the main group to be superior in terms of muscle strength (according to the MRC scale (visit 4 - 3.3±5.1 vs 2.1±2.2, p=0.051), level of vital activity (according to the ShRM - visit 5 - 2.9±0.7 vs 3.3±0.6, p=0.053). A statistically significant increase in the level of mobility of patients in the group using the drug Mexidol was proven compared to the control group (the difference in the Rivermead index at the 5th visit was 10.3±2.8 and 8.0±2.8, respectively, p=0.006), the average increase in the Rivermead index by visit 5 (5.4±2.1 vs 3.4±1.6, p<0.001). A decrease in intensive care aftereffects syndrome (ITS) scores was detected in both groups; a statistically significant decrease in the severity of ITS in relation to the previous visit was detected only in the group using the drug Mexidol (p<0.001). In the main group, the best indicators of the dynamics of systolic cerebral blood flow velocity and overshoot coefficient were also determined, compared to the control group. There were no adverse events recorded in the study. CONCLUSION: A positive modulating effect of Mexidol has been demonstrated in terms of accelerating the restoration of tolerance to cognitive loads, improving the psycho-emotional background by reducing symptoms of anxiety and depression, and secondary improving the results of motor rehabilitation in the early recovery period in patients who have undergone ACI, including those with manifestations of PIT syndrome. During the study, no adverse events were recorded, as well as significant differences in vital functions in the study groups, which indicates comparable safety of therapy in the control and main groups.


Subject(s)
Picolines , Humans , Picolines/therapeutic use , Picolines/administration & dosage , Male , Female , Middle Aged , Adult , Prospective Studies , Treatment Outcome , Stroke/drug therapy , Stroke/complications , Stroke/physiopathology , Brain Injuries, Traumatic/rehabilitation , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/complications , Stroke Rehabilitation/methods , Aged , Anxiety/drug therapy , Anxiety/etiology
10.
Molecules ; 29(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38675525

ABSTRACT

Traumatic brain injury (TBI) leads to structural damage in the brain, and is one of the major causes of disability and death in the world. Herein, we developed a composite injectable hydrogel (HA/Gel) composed of hyaluronic acid (HA) and gelatin (Gel), loaded with vascular endothelial growth factor (VEGF) and salvianolic acid B (SAB) for treatment of TBI. The HA/Gel hydrogels were formed by the coupling of phenol-rich tyramine-modified HA (HA-TA) and tyramine-modified Gel (Gel-TA) catalyzed by horseradish peroxidase (HRP) in the presence of hydrogen peroxide (H2O2). SEM results showed that HA/Gel hydrogel had a porous structure. Rheological test results showed that the hydrogel possessed appropriate rheological properties, and UV spectrophotometry results showed that the hydrogel exhibited excellent SAB release performance. The results of LIVE/DEAD staining, CCK-8 and Phalloidin/DAPI fluorescence staining showed that the HA/Gel hydrogel possessed good cell biocompatibility. Moreover, the hydrogels loaded with SAB and VEGF (HA/Gel/SAB/VEGF) could effectively promote the proliferation of bone marrow mesenchymal stem cells (BMSCs). In addition, the results of H&E staining, CD31 and α-SMA immunofluorescence staining showed that the HA/Gel/SAB/VEGF hydrogel possessed good in vivo biocompatibility and pro-angiogenic ability. Furthermore, immunohistochemical results showed that the injection of HA/Gel/SAB/VEGF hydrogel to the injury site could effectively reduce the volume of defective tissues in traumatic brain injured mice. Our results suggest that the injection of HA/Gel hydrogel loaded with SAB and VEGF might provide a new approach for therapeutic brain tissue repair after traumatic brain injury.


Subject(s)
Benzofurans , Brain Injuries, Traumatic , Depsides , Gelatin , Hyaluronic Acid , Hydrogels , Vascular Endothelial Growth Factor A , Animals , Hydrogels/chemistry , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Gelatin/chemistry , Hyaluronic Acid/chemistry , Mice , Vascular Endothelial Growth Factor A/metabolism , Benzofurans/chemistry , Benzofurans/pharmacology , Benzofurans/administration & dosage , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Disease Models, Animal , Male , Cell Proliferation/drug effects
11.
Neurochem Int ; 176: 105742, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641028

ABSTRACT

Traumatic brain injury (TBI) remains a major health concern which causes long-term neurological disability particularly in war veterans, athletes and young adults. In spite of intense clinical and research investigations, there is no effective therapy to cease the pathogenesis of the disease. It is believed that axonal injury during TBI is potentiated by neuroinflammation and demyelination and/or failure to remyelination. This study highlights the use of naturally available cinnamein, also chemically known as benzyl cinnamate, in inhibiting neuroinflammation, promoting remyelination and combating the disease process of controlled cortical impact (CCI)-induced TBI in mice. Oral delivery of cinnamein through gavage brought down the activation of microglia and astrocytes to decrease the expression of inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in hippocampus and cortex of TBI mice. Cinnamein treatment also stimulated remyelination in TBI mice as revealed by PLP and A2B5 double-labeling, luxol fast blue (LFB) staining and axonal double-labeling for neurofilament and MBP. Furthermore, oral cinnamein reduced the size of lesion cavity in the brain, improved locomotor functions and restored memory and learning in TBI mice. These results suggest a new neuroprotective property of cinnamein that may be valuable in the treatment of TBI.


Subject(s)
Brain Injuries, Traumatic , Disease Models, Animal , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Mice , Male , Mice, Inbred C57BL , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
12.
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
14.
Iran Biomed J ; 28(2&3): 113-9, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38562043

ABSTRACT

Background: Traumatic brain injury or TBI can underlie epilepsy. Prevention of PTE has been of great interest to scientists. Given the antiepileptic, antioxidant and anti-inflammatory activities of curcumin, we examined whether this compound can affect epileptogenesis in rats after TBI. Methods: Curcumin was injected once a day for two weeks. TBI was induced in the temporal cortex of anesthetized rats using a CCI device. One day after TBI, PTZ, 35 mg/kg, was injected i.p. every other day until manifestation of generalized seizures. The number of PTZ injections was then recorded. Moreover, the extent of cortical and hippocampal IL-1ß and GFAP expression in the epileptic rats were measured by Western blot analysis. Results: Curcumin 50 and 150 mg/kg prevented the development of kindling, wherase TBI accelerated the rate of kindling. Curcumin 20 mg/kg prohibited kindling facilitation by TBI, and reduced the expression of IL-1ß and GFAP induced by TBI. Conclusion: Curcumin can stop the acceleration of epileptogenesis after TBI in rats. Inhibiting hippocampal and cortical overexpression of IL-1ß and GFAP seems to be involved in this activity.


Subject(s)
Brain Injuries, Traumatic , Curcumin , Epilepsy , Glial Fibrillary Acidic Protein , Hippocampus , Interleukin-1beta , Kindling, Neurologic , Curcumin/pharmacology , Curcumin/therapeutic use , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/complications , Interleukin-1beta/metabolism , Male , Epilepsy/drug therapy , Glial Fibrillary Acidic Protein/metabolism , Kindling, Neurologic/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Rats, Sprague-Dawley , Seizures/drug therapy
15.
Exp Neurol ; 377: 114795, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657855

ABSTRACT

Clinical studies have shown that traumatic brain injury (TBI) increases the onset of Parkinson's disease (PD) in later life by >50%. Oxidative stress, endoplasmic reticulum (ER) stress, and inflammation are the major drivers of both TBI and PD pathologies. We presently evaluated if curtailing oxidative stress and ER stress concomitantly using a combination of apocynin and tert-butylhydroquinone and salubrinal during the acute stage after TBI in mice reduces the severity of late-onset PD-like pathology. The effect of multiple low doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on post-TBI neurodegeneration was also evaluated. The combo therapy elevated the level of phosphorylation at serine 129 (pS129) of α-Syn in the pericontusional cortex of male mice at 72 h post-TBI. Motor and cognitive deficits induced by TBI lasted at least 3 months and the combo therapy curtailed these deficits in both sexes. At 3 months post-TBI, male mice given combo therapy exhibited significantly lesser α-Syn aggregates in the SN and higher TH+ cells in the SNpc, compared to vehicle control. However, the aggregate number was not significantly different between groups of female mice. Moreover, TBI-induced loss of TH+ cells was negligible in female mice irrespective of treatment. The MPTP treatment aggravated PD-like pathology in male mice but had a negligible effect on the loss of TH+ cells in female mice. Thus, the present study indicates that mitigation of TBI-induced oxidative stress and ER stress at the acute stage could potentially reduce the risk of post-TBI PD-like pathology at least in male mice, plausibly by elevating pS129-α-Syn level.


Subject(s)
Antioxidants , Brain Injuries, Traumatic , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , Animals , Male , Mice , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/drug therapy , Female , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Phosphorylation/drug effects , Antioxidants/pharmacology , Sex Characteristics , Acetophenones/pharmacology , Acetophenones/therapeutic use , Acetophenones/administration & dosage , Thiourea/analogs & derivatives , Thiourea/pharmacology , Thiourea/therapeutic use , Thiourea/administration & dosage , Serine/metabolism , Hydroquinones/pharmacology , Hydroquinones/administration & dosage , Hydroquinones/therapeutic use , Drug Therapy, Combination , Oxidative Stress/drug effects
16.
Phytomedicine ; 129: 155566, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38565001

ABSTRACT

BACKGROUND: Xuefu Zhuyu decoction (XFZYD) is a traditional Chinese herbal formula known for its ability to eliminate blood stasis and improve blood circulation, providing neuroprotection against severe traumatic brain injury (sTBI). However, the underlying mechanism is still unclear. PURPOSE: We aim to investigate the neuroprotective effects of XFZYD in sTBI from a novel mechanistic perspective of miRNA-mRNA. Additionally, we sought to elucidate a potential specific mechanism by integrating transcriptomics, bioinformatics, and conducting both in vitro and in vivo experiments. METHODS: The sTBI rat model was established, and the rats were treated with XFZYD for 14 days. The neuroprotective effects of XFZYD were evaluated using a modified neurological severity score, hematoxylin and eosin staining, as well as Nissl staining. The anti-inflammatory effects of XFZYD were explored using quantitative real-time PCR (qRT-PCR), Western blot analysis, and immunofluorescence. Next, miRNA sequencing of the hippocampus was performed to determine which miRNAs were differentially expressed. Subsequently, qRT-PCR was used to validate the differentially expressed miRNAs. Target core mRNAs were determined using various methods, including miRNA prediction targets, mRNA sequencing, miRNA-mRNA network, and protein-protein interaction (PPI) analysis. The miRNA/mRNA regulatory axis were verified through qRT-PCR or Western blot analysis. Finally, morphological changes in the neural synapses were observed using transmission electron microscopy and immunofluorescence. RESULTS: XFZYD exhibited significant neuroprotective and anti-inflammatory effects on subacute sTBI rats' hippocampus. The analyses of miRNA/mRNA sequences combined with the PPI network revealed that the therapeutic effects of XFZYD on sTBI were associated with the regulation of the rno-miR-191a-5p/BDNF axis. Subsequently, qRT-PCR and Western blot analysis confirmed XFZYD reversed the decrease of BDNF and TrkB in the hippocampus caused by sTBI. Additionally, XFZYD treatment potentially increased the number of synaptic connections, and the expression of the synapse-related protein PSD95, axon-related protein GAP43 and neuron-specific protein TUBB3. CONCLUSIONS: XFZYD exerts neuroprotective effects by promoting hippocampal synaptic remodeling and improving cognition during the subacute phase of sTBI through downregulating of rno-miR-191a-5p/BDNF axis, further activating BDNF-TrkB signaling.


Subject(s)
Brain Injuries, Traumatic , Brain-Derived Neurotrophic Factor , Drugs, Chinese Herbal , Hippocampus , MicroRNAs , Neuronal Plasticity , Neuroprotective Agents , Rats, Sprague-Dawley , Animals , MicroRNAs/metabolism , Brain Injuries, Traumatic/drug therapy , Drugs, Chinese Herbal/pharmacology , Neuronal Plasticity/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Male , Rats , Neuroprotective Agents/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal , Receptor, trkB/metabolism
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.
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
19.
Drug Des Devel Ther ; 18: 1175-1188, 2024.
Article in English | MEDLINE | ID: mdl-38645986

ABSTRACT

Purpose: Many herbs can promote neurological recovery following traumatic brain injury (TBI). There must lie a shared mechanism behind the common effectiveness. We aimed to explore the key therapeutic targets for TBI based on the common effectiveness of the medicinal plants. Material and methods: The TBI-effective herbs were retrieved from the literature as imputes of network pharmacology. Then, the active ingredients in at least two herbs were screened out as common components. The hub targets of all active compounds were identified through Cytohubba. Next, AutoDock vina was used to rank the common compound-hub target interactions by molecular docking. A highly scored compound-target pair was selected for in vivo validation. Results: We enrolled sixteen TBI-effective medicinal herbs and screened out twenty-one common compounds, such as luteolin. Ten hub targets were recognized according to the topology of the protein-protein interaction network of targets, including epidermal growth factor receptor (EGFR). Molecular docking analysis suggested that luteolin could bind strongly to the active pocket of EGFR. Administration of luteolin or the selective EGFR inhibitor AZD3759 to TBI mice promoted the recovery of body weight and neurological function, reduced astrocyte activation and EGFR expression, decreased chondroitin sulfate proteoglycans deposition, and upregulated GAP43 levels in the cortex. The effects were similar to those when treated with the selective EGFR inhibitor. Conclusion: The common effectiveness-based, common target screening strategy suggests that inhibition of EGFR can be an effective therapy for TBI. This strategy can be applied to discover core targets and therapeutic compounds in other diseases.


Subject(s)
Brain Injuries, Traumatic , Molecular Docking Simulation , Network Pharmacology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Animals , Mice , Plants, Medicinal/chemistry , Male , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Luteolin/pharmacology , Luteolin/chemistry , Mice, Inbred C57BL , Humans
20.
Sci Rep ; 14(1): 7626, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38561445

ABSTRACT

This study explored the application of generative pre-trained transformer (GPT) agents based on medical guidelines using large language model (LLM) technology for traumatic brain injury (TBI) rehabilitation-related questions. To assess the effectiveness of multiple agents (GPT-agents) created using GPT-4, a comparison was conducted using direct GPT-4 as the control group (GPT-4). The GPT-agents comprised multiple agents with distinct functions, including "Medical Guideline Classification", "Question Retrieval", "Matching Evaluation", "Intelligent Question Answering (QA)", and "Results Evaluation and Source Citation". Brain rehabilitation questions were selected from the doctor-patient Q&A database for assessment. The primary endpoint was a better answer. The secondary endpoints were accuracy, completeness, explainability, and empathy. Thirty questions were answered; overall GPT-agents took substantially longer and more words to respond than GPT-4 (time: 54.05 vs. 9.66 s, words: 371 vs. 57). However, GPT-agents provided superior answers in more cases compared to GPT-4 (66.7 vs. 33.3%). GPT-Agents surpassed GPT-4 in accuracy evaluation (3.8 ± 1.02 vs. 3.2 ± 0.96, p = 0.0234). No difference in incomplete answers was found (2 ± 0.87 vs. 1.7 ± 0.79, p = 0.213). However, in terms of explainability (2.79 ± 0.45 vs. 07 ± 0.52, p < 0.001) and empathy (2.63 ± 0.57 vs. 1.08 ± 0.51, p < 0.001) evaluation, the GPT-agents performed notably better. Based on medical guidelines, GPT-agents enhanced the accuracy and empathy of responses to TBI rehabilitation questions. This study provides guideline references and demonstrates improved clinical explainability. However, further validation through multicenter trials in a clinical setting is necessary. This study offers practical insights and establishes groundwork for the potential theoretical integration of LLM-agents medicine.


Subject(s)
Brain Injuries, Traumatic , Humans , Brain Injuries, Traumatic/drug therapy , Brain , Databases, Factual , Electric Power Supplies , Empathy
SELECTION OF CITATIONS
SEARCH DETAIL