Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 155
1.
Sci Rep ; 14(1): 6027, 2024 03 12.
Article En | MEDLINE | ID: mdl-38472318

We have previously shown that the pro-oxidative aldehyde acrolein is a critical factor in MS pathology. In this study, we found that the acrolein scavenger hydralazine (HZ), when applied from the day of induction, can suppress acrolein and alleviate motor and sensory deficits in a mouse experimental autoimmune encephalomyelitis (EAE) model. Furthermore, we also demonstrated that HZ can alleviate motor deficits when applied after the emergence of MS symptoms, making potential anti-acrolein treatment a more clinically relevant strategy. In addition, HZ can reduce both acrolein and MPO, suggesting a connection between acrolein and inflammation. We also found that in addition to HZ, phenelzine (PZ), a structurally distinct acrolein scavenger, can mitigate motor deficits in EAE when applied from the day of induction. This suggests that the likely chief factor of neuroprotection offered by these two structurally distinct acrolein scavengers in EAE is their common feature of acrolein neutralization. Finally, up-and-down regulation of the function of aldehyde dehydrogenase 2 (ALDH2) in EAE mice using either a pharmacological or genetic strategy led to correspondent motor and sensory changes. This data indicates a potential key role of ALDH2 in influencing acrolein levels, oxidative stress, inflammation, and behavior in EAE. These findings further consolidate the critical role of aldehydes in the pathology of EAE and its mechanisms of regulation. This is expected to reinforce and expand the possible therapeutic targets of anti-aldehyde treatment to achieve neuroprotection through both endogenous and exogenous manners.


Acrolein , Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , Acrolein/pharmacology , Encephalomyelitis, Autoimmune, Experimental/pathology , Neuroprotection , Phenelzine/pharmacology , Aldehydes , Inflammation/pathology , Mice, Inbred C57BL
2.
Ecotoxicol Environ Saf ; 275: 116241, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38522287

Iron overload occurs due to excessive iron intake compared to the body's demand, leading to iron deposition and impairment of multiple organ functions. Our previous study demonstrated that chronic oral administration of ferric citrate (FC) caused colonic inflammatory injury. However, the precise mechanism underlying this inflammatory response remains unclear. The current study aims to investigate the mechanism by which iron overload induced by FC exposure leads to colonic inflammation. To accomplish this, mice were orally exposed to three different concentrations of FC (71 mg/kg/bw (L), 143 mg/kg/bw (M) and 286 mg/kg/bw (H)) for continuous 16 weeks, with the control group receiving ultrapure water (C). Exposure to FC caused disturbances in the excretory system, altered colonic flora alpha diversity, and enriched pathogenic bacteria, such as Mucispirillum, Helicobacter, Desulfovibrio, and Shigella. These changes led to structural disorders of the colonic flora and an inflammatory response phenotype characterized by inflammatory cells infiltration, atrophy of intestinal glands, and irregular thickening of the intestinal wall. Mechanistic studies revealed that FC-exposure activated the NF-κB signaling pathway by up-regulating TLR4, MyD88, and NF-κB mRNA levels and protein expression. This activation resulted in increased production of pro-inflammatory cytokines, further contributing to the colonic inflammation. Additionally, in vitro experiments in SW480 cells confirmed the activation of NF-κB signaling pathway by FC exposure, consistent with the in vivo findings. The significance of this study lies in its elucidation of the mechanism by which iron overload caused by FC exposure leads to colonic inflammation. By identifying the role of pathogenic bacteria and the NF-κB signaling pathway, this study could potentially offer a crucial theoretical foundation for the research on iron overload, as well as provide valuable insights for clinical iron supplementation.


Ferric Compounds , Iron Overload , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Inflammation/chemically induced , Inflammation/pathology , Iron Overload/pathology , Iron/metabolism
3.
Tissue Eng Regen Med ; 21(3): 437-453, 2024 Apr.
Article En | MEDLINE | ID: mdl-38308742

BACKGROUND: Previous investigations have shown that local application of nanoparticles presenting the carbohydrate moiety galactose-α-1,3-galactose (α-gal epitopes) enhance wound healing by activating the complement system and recruiting pro-healing macrophages to the injury site. Our companion in vitro paper suggest α-gal epitopes can similarly recruit and polarize human microglia toward a pro-healing phenotype. In this continuation study, we investigate the in vivo implications of α-gal nanoparticle administration directly to the injured spinal cord. METHODS: α-Gal knock-out (KO) mice subjected to spinal cord crush were injected either with saline (control) or with α-gal nanoparticles immediately following injury. Animals were assessed longitudinally with neurobehavioral and histological endpoints. RESULTS: Mice injected with α-gal nanoparticles showed increased recruitment of anti-inflammatory macrophages to the injection site in conjunction with increased production of anti-inflammatory markers and a reduction in apoptosis. Further, the treated group showed increased axonal infiltration into the lesion, a reduction in reactive astrocyte populations and increased angiogenesis. These results translated into improved sensorimotor metrics versus the control group. CONCLUSIONS: Application of α-gal nanoparticles after spinal cord injury (SCI) induces a pro-healing inflammatory response resulting in neuroprotection, improved axonal ingrowth into the lesion and enhanced sensorimotor recovery. The data shows α-gal nanoparticles may be a promising avenue for further study in CNS trauma.


Nanoparticles , Spinal Cord Injuries , Mice , Humans , Animals , Galactose/therapeutic use , Spinal Cord Injuries/drug therapy , Mice, Knockout , Anti-Inflammatory Agents , Epitopes/therapeutic use , Immunomodulation
4.
Tissue Eng Regen Med ; 21(3): 409-419, 2024 Apr.
Article En | MEDLINE | ID: mdl-38099990

BACKGROUND: Macrophages and microglia play critical roles after spinal cord injury (SCI), with the pro-healing, anti-inflammatory (M2) subtype being implicated in tissue repair. We hypothesize that promoting this phenotype within the post-injured cord microenvironment may provide beneficial effects for mitigating tissue damage. As a proof of concept, we propose the use of nanoparticles incorporating the carbohydrate antigen, galactose-α-1,3-galactose (α-gal epitope) as an immunomodulator to transition human microglia (HMC3) cells toward a pro-healing state. METHODS: Quiescent HMC3 cells were acutely exposed to α-gal nanoparticles in the presence of human serum and subsequently characterized for changes in cell shape, expression of anti or pro-inflammatory markers, and secretion of phenotype-specific cytokines. RESULTS: HMC3 cells treated with serum activated α-gal nanoparticles exhibited rapid enlargement and shape change in addition to expressing CD68. Moreover, these activated cells showed increased expression of anti-inflammatory markers like Arginase-1 and CD206 without increasing production of pro-inflammatory cytokines TNF-α or IL-6. CONCLUSION: This study is the first to show that resting human microglia exposed to a complex of α-gal nanoparticles and anti-Gal (from human serum) can be activated and polarized toward a putative M2 state. The data suggests that α-gal nanoparticles may have therapeutic relevance to the CNS microenvironment, in both recruiting and polarizing macrophages/microglia at the application site. The immunomodulatory activity of these α-gal nanoparticles post-SCI is further described in the companion work (Part II).


Nanoparticles , Spinal Cord Injuries , Humans , Microglia/metabolism , Galactose/metabolism , Galactose/therapeutic use , Cytokines/metabolism , Anti-Inflammatory Agents
5.
Int J Mol Sci ; 24(17)2023 Aug 30.
Article En | MEDLINE | ID: mdl-37686246

In recent years, olfactory dysfunction has attracted increasingly more attention as a hallmark symptom of neurodegenerative diseases (ND). Deeply understanding the molecular basis underlying the development of the olfactory bulb (OB) will provide important insights for ND studies and treatments. Now, with a genetic knockout mouse model, we show that TRIM67, a new member of the tripartite motif (TRIM) protein family, plays an important role in regulating the proliferation and development of mitral cells in the OB. TRIM67 is abundantly expressed in the mitral cell layer of the OB. The genetic deletion of TRIM67 in mice leads to excessive proliferation of mitral cells in the OB and defects in its synaptic development, resulting in reduced olfactory function in mice. Finally, we show that TRIM67 may achieve its effect on mitral cells by regulating the Semaphorin 7A/Plexin C1 (Sema7A/PlxnC1) signaling pathway.


Olfactory Bulb , Smell , Animals , Mice , Homeostasis , Gene Deletion , Tripartite Motif Proteins , Cytoskeletal Proteins
6.
Lab Chip ; 23(15): 3388-3404, 2023 07 25.
Article En | MEDLINE | ID: mdl-37337817

Trauma-induced Alzheimer's disease (AD) is rapidly emerging as a major consequence of traumatic brain injuries (TBI), with devastating social and economic impacts. Unfortunately, few treatment options are currently available due to a limited understanding of the underlying mechanisms. A clinically-relevant, in vitro experimental model that emulates in vivo scenarios with high levels of spatial and temporal resolution is critical for demystifying the pathways of post-TBI AD. Using a unique, recently established "TBI-on-a-chip" system with murine cortical networks, we demonstrate the correlative elevation of oxidative stress (acrolein), inflammation (TNF-α), and Aß42 aggregation, with concomitant reduction of neuronal network electrical activity post-concussive impact. These findings confirm that TBI-on-a-chip could provide a novel paradigm to supplement in vivo studies of trauma, while simultaneously validating the interaction of these alleged, key-pathological factors in post-TBI AD development. Specifically, we have shown that acrolein, acting as a diffusive factor of secondary injury, is both critical and sufficient in promoting inflammation (TNF-α) and Aß42 aggregation, two known contributors of AD pathogenesis. Furthermore, using a cell-free preparation with TBI-on-a-chip, we have confirmed that both force and acrolein can independently and directly stimulate the aggregation of purified Aß42, highlighting the key capabilities of primary and secondary injury mechanisms towards inducing Aß42 aggregation, independently and synergistically. In addition to morphological and biochemical assessment, we also demonstrate parallel monitoring of neuronal network activity, further validating the chief pathological role of acrolein in not only inflicting biochemical abnormalities, but also functional deficits in neuronal networks. In conclusion, through this line of investigations, we have shown that by recapitulating clinically-relevant events, the TBI-on-a-chip device is capable of quantitatively characterizing parallel force-dependent increases in oxidative stress, inflammation, protein aggregation, and network activity, offering a unique platform for mechanistic investigations of post-TBI AD, and trauma-induced neuronal injury in general. It is expected that this model could provide crucial insights into pathological mechanisms which will be critical in developing novel, effective diagnostics and treatment strategies that significantly benefit TBI victims.


Alzheimer Disease , Brain Injuries, Traumatic , Mice , Animals , Amyloid beta-Peptides , Acrolein , Tumor Necrosis Factor-alpha , Brain Injuries, Traumatic/pathology , Lab-On-A-Chip Devices , Inflammation/complications
8.
Mol Neurobiol ; 60(5): 2937-2953, 2023 May.
Article En | MEDLINE | ID: mdl-36750527

Spinal cord injury is a severely debilitating condition affecting a significant population in the USA. Spinal cord injury patients often have increased risk of developing persistent neuropathic pain and other neurodegenerative conditions beyond the primary lesion center later in their life. The molecular mechanism conferring to the "latent" damages at distal tissues, however, remains elusive. Here, we studied molecular changes conferring abnormal functionality at distal spinal cord (T12) beyond the lesion center (T10) by combining next-generation sequencing (RNA- and bisulfite sequencing), super-resolution microscopy, and immunofluorescence staining at 7 days post injury. We observed significant transcriptomic changes primarily enriched in neuroinflammation and synaptogenesis associated pathways. Transcription factors (TFs) that regulate neurogenesis and neuron plasticity, including Egr1, Klf4, and Myc, are significantly upregulated. Along with global changes in chromatin arrangements and DNA methylation, including 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC), bisulfite sequencing further reveals the involvement of DNA methylation changes in regulating cytokine, growth factor, and ion channel expression. Collectively, our results pave the way towards understanding transcriptomic and epigenomic mechanism in conferring long-term disease risks at distal tissues away from the primary lesion center and shed light on potential molecular targets that govern the regulatory mechanism at distal spinal cord tissues.


Contusions , Spinal Cord Injuries , Rats , Animals , Epigenesis, Genetic , Transcriptome/genetics , Epigenomics/methods , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , DNA Methylation/genetics , Spinal Cord/pathology
9.
Ecotoxicol Environ Saf ; 249: 114364, 2023 Jan 01.
Article En | MEDLINE | ID: mdl-36508806

Ferric citrate (FC) has been used as an iron fortifier and nutritional supplement, which is reported to induce colitis in rats, however the underlying mechanism remains to be elucidated. We performed a 16-week study of FC in male healthy C57BL/6 mice (nine-month-old) with oral administration of Ctr (0.9 % NaCl), 1.25 % FC (71 mg/kg/bw), 2.5 % FC (143 mg/kg/bw) and 5 % FC (286 mg/kg/bw). FC-exposure resulted in colon iron accumulation, histological alteration and reduce antioxidant enzyme activities, such as glutathione (GSH), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and total antioxidant capacity (T-AOC), together with enhanced lipid peroxidation level, including malondialdehyde (MDA) level and 4-Hydroxynonenal (4-HNE) protein expression. Exposure to FC was associated with upregulated levels of the interleukin (IL)- 6, IL-1ß, IL-18, IL-8 and tumor necrosis factor α (TNF-α), while down-regulated levels of IL-4 and IL-10. Exposure to FC was positively associated with the mRNA and protein expressions of cysteine-aspartic proteases (Caspase)- 9, Caspase-3, Bcl-2-associated X protein (Bax), while negatively associated with B-cell lymphoma 2 (Bcl2) in mitochondrial apoptosis signaling pathway. FC-exposure changed the diversity and composition of gut microbes. Additionally, the serum lipopolysaccharide (LPS) contents increased in FC-exposed groups when compared with the control group, while the expression of colonic tight junction proteins (TJPs), such as Claudin-1 and Occludin were decreased. These findings indicate that the colonic mucosal injury induced by FC-exposure are associated with oxidative stress generation, inflammation response and cell apoptosis, as well as the changes in gut microbes diversity and composition.


Apoptosis , Colon , Ferric Compounds , Food, Fortified , Gastrointestinal Microbiome , Inflammation , Oxidative Stress , Animals , Male , Mice , Rats , Apoptosis/drug effects , Colon/drug effects , Colon/metabolism , Ferric Compounds/toxicity , Food, Fortified/toxicity , Gastrointestinal Microbiome/drug effects , Glutathione/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Intestinal Mucosa/drug effects , Iron/metabolism , Mice, Inbred C57BL , Superoxide Dismutase/metabolism
10.
Eur J Pharmacol ; 940: 175474, 2023 Feb 05.
Article En | MEDLINE | ID: mdl-36549500

Dysregulation of neuronal development may cause neurodevelopmental disorders. However, how to regulate embryonic neuronal development and whether this regulation can be medical interrupted are largely unknown. This study aimed to investigate whether and how andrographolide (ANP) regulates embryonic neuronal development. The pregnant mice at embryonic day 10.5 (E10.5) were administrated with ANP, and the embryonic brains were harvested at E17.5 or E18.5. Immunofluorescence (IF), Immunohistochemistry (IHC) performed to determine whether ANP is critical in regulating neuronal development. Real-time quantitative PCR, western blotting, cell counting kit-8 assay, Flow Cytometry assay, Boyden Chamber Migration assay carried out to evaluate whether ANP regulates neuronal proliferation and migration. Protein-protein interaction, CO-immunoprecipitation and IF staining carried out to evaluate whether ANP regulates the interaction between PFKFB3, NeuN and TBR1. Knockdown or overexpression of PFKFB3 by adenovirus infection were used to determine whether ANP inhibits neuronal development through PFKFB3 mediated glycolytic pathway. Our data indicated that ANP inhibited the maturation of embryonic neurons characterized by suppressing neuronal proliferation and migration. ANP regulated the interaction between PFKFB3, NeuN, and TBR1. Knockdown of PFKFB3 aggravated ANP mediated inhibition of neuronal proliferation and migration, while overexpression of PFKFB3 attenuated ANP mediated neuronal developmental suppression. In summary, ANP suppressed the expression of PFKFB3, and interrupted the interaction between TRB1 and NeuN, resulting in suppressing neuronal proliferation, migration and maturation and eventually inhibiting murine embryonic neuronal development.


Diterpenes , Phosphofructokinase-2 , Pregnancy , Female , Mice , Animals , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Phosphoric Monoester Hydrolases/metabolism , Diterpenes/pharmacology , Glycolysis , Cell Proliferation
11.
J Spinal Cord Med ; 46(3): 508-520, 2023 05.
Article En | MEDLINE | ID: mdl-33890843

Background: Spinal cord injuries cause great damage to the central nervous system as well as the peripheral vasculature. While treatments for spinal cord injury typically focus on the spine itself, improvements in the function of the peripheral vasculature after spinal cord injury have shown to improve overall neurological recovery.Objective: This study focused on the use of near-infrared spectroscopy (NIRS) as a mode to monitor cerebral and peripheral vascular condition non-invasively during the recovery process.Design: Animal research study.Methods: Rats underwent spinal contusion or sham injury and relative concentrations of de-/oxyhemoglobin (Δ[HbO]/Δ[Hb]) over time were measured over the cerebral, spinal, and pedal regions via NIRS. Correlational relationships across the body were determined. Rats received 1 NIRS measurement before injury and 3 after injury: 4, 7, and 14 days post.Results: Correlational relationships between signals across the body, between animals with and without spinal cord injury, indicate that NIRS was able to detect patterns of vascular change in the spine and the periphery occurring secondary to spinal cord injury and evolving during subsequent recovery. Additionally, NIRS determined an overall correlational decrease within the central nervous system, between spinal and cerebral measurements.Conclusion: NIRS was able to closely reflect physiologic changes in the rat during recovery, demonstrating a promising method to monitor whole body hemodynamics after spinal cord injury.


Contusions , Spinal Cord Injuries , Rats , Animals , Spinal Cord Injuries/therapy , Spinal Cord , Spectroscopy, Near-Infrared/methods , Central Nervous System
12.
Magn Reson Med ; 89(2): 508-521, 2023 02.
Article En | MEDLINE | ID: mdl-36161728

PURPOSE: This study aimed to develop a new 3D dual-echo rosette k-space trajectory, specifically designed for UTE MRI applications. The imaging of the ultra-short transverse relaxation time (uT2 ) of brain was acquired to test the performance of the proposed UTE sequence. THEORY AND METHODS: The rosette trajectory was developed based on rotations of a "petal-like" pattern in the kx -ky plane, with oscillated extensions in the kz -direction for 3D coverage. Five healthy volunteers underwent 10 dual-echo 3D rosette UTE scans with various TEs. Dual-exponential complex model fitting was performed on the magnitude data to separate uT2 signals, with the output of uT2 fraction, uT2 value, and long-T2 value. RESULTS: The 3D rosette dual-echo UTE sequence showed better performance than a 3D radial UTE acquisition. More significant signal intensity decay in white matter than gray matter was observed along with the TEs. The white matter regions had higher uT2 fraction values than gray matter (10.9% ± 1.9% vs. 5.7% ± 2.4%). The uT2 value was approximately 0.10 ms in white matter . CONCLUSION: The higher uT2 fraction value in white matter compared to gray matter demonstrated the ability of the proposed sequence to capture rapidly decaying signals.


Magnetic Resonance Imaging , White Matter , Humans , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Gray Matter/diagnostic imaging , Healthy Volunteers , Imaging, Three-Dimensional
13.
Neurotrauma Rep ; 3(1): 501-510, 2022.
Article En | MEDLINE | ID: mdl-36479362

Autonomic dysreflexia (AD) frequently occurs in persons with spinal cord injuries (SCIs) above the T6 level triggered by different stimuli below the level of injury. If improperly managed, AD can have severe clinical consequences, even possibly leading to death. Existing techniques for AD detection are time-consuming, obtrusive, lack automated detection capabilities, and have low temporal resolution. Therefore, a non-invasive, multi-modal wearable diagnostic tool was developed to quantitatively characterize and distinguish unique signatures of AD. Electrocardiography and novel skin nerve activity (skNA) sensors with neural networks were used to detect temporal changes in the sympathetic and vagal systems in rats with SCI. Clinically established metrics of AD were used to verify the onset of AD. Five physiological features reflecting different metrics of sympathetic and vagal activity were used to characterize signatures of AD. An increase in sympathetic activity, followed by a lagged increase in vagal activity during the onset of AD, was observed after inducing AD. This unique signature response was used to train a neural network to detect the onset of AD with an accuracy of 93.4%. The model also had a 79% accuracy in distinguishing between sympathetic hyperactivity reactions attributable to different sympathetic stressors above and below the level of injury. These neural networks have not been used in previous work to detect the onset of AD. The system could serve as a complementary non-invasive tool to the clinically accepted gold standard, allowing an improved management of AD in persons with SCI.

14.
Front Immunol ; 13: 952852, 2022.
Article En | MEDLINE | ID: mdl-36059492

There are no licensed therapeutics or vaccines available against porcine delta coronavirus (PDCoV) to eliminate its potential for congenital disease. In the absence of effective treatments, it has led to significant economic losses in the swine industry worldwide. Similar to the current coronavirus disease 2019 (COVID-19) pandemic, PDCoV is trans-species transmissible and there is still a large desert for scientific exploration. We have reported that selenomethionine (SeMet) has potent antiviral activity against PDCoV. Here, we systematically investigated the endogenous immune mechanism of SeMet and found that STAT3/miR-125b-5p-1/HK2 signalling is essential for the exertion of SeMet anti-PDCoV replication function. Meanwhile, HK2, a key rate-limiting enzyme of the glycolytic pathway, was able to control PDCoV replication in LLC-PK1 cells, suggesting a strategy for viruses to evade innate immunity using glucose metabolism pathways. Overall, based on the ability of selenomethionine to control PDCoV infection and transmission, we provide a molecular basis for the development of new therapeutic approaches.


COVID-19 , MicroRNAs , Swine Diseases , Animals , LLC-PK1 Cells , Selenomethionine/pharmacology , Swine
15.
Front Immunol ; 13: 972499, 2022.
Article En | MEDLINE | ID: mdl-36081520

Porcine Deltacoronavirus (PDCoV), an enveloped positive-strand RNA virus that causes respiratory and gastrointestinal diseases, is widely spread worldwide, but there is no effective drug or vaccine against it. This study investigated the optimal Selenium Nano-Particles (SeNPs) addition concentration (2 - 10 µg/mL) and the mechanism of PDCoV effect on ST (Swine Testis) cell apoptosis, the antagonistic effect of SeNPs on PDCoV. The results indicated that 4 µg/mL SeNPs significantly decreased PDCoV replication on ST cells. SeNPs relieved PDCoV-induced mitochondrial division and antagonized PDCoV-induced apoptosis via decreasing Cyt C release and Caspase 9 and Caspase 3 activation. The above results provided an idea and experimental basis associated with anti-PDCoV drug development and clinical use.


Coronavirus Infections , Coronavirus , Selenium , Swine Diseases , Animals , Apoptosis , Coronavirus/physiology , Male , Mitochondrial Dynamics , Swine
16.
Int J Mol Sci ; 23(16)2022 Aug 19.
Article En | MEDLINE | ID: mdl-36012632

Colon cancer is a common malignant tumor of the digestive tract, and it is considered among the biggest killers. Scientific and reasonable treatments can effectively improve the survival rate of patients if performed in the early stages. Polyphyllin I (PPI), a pennogenyl saponin isolated from Paris polyphylla var. yunnanensis, has exhibited strong anti-cancer activities in previous studies. Here, we report that PPI exhibits a cytotoxic effect on colon cancer cells. PPI suppressed cell viability and induced autophagic cell death in SW480 cells after 12 and 24 h, with the IC50 values 4.9 ± 0.1 µmol/L and 3.5 ± 0.2 µmol/L, respectively. Furthermore, we found PPI induced time-concentration-dependent autophagy and apoptosis in SW480 cells. In addition, down-regulated AKT/mTOR activity was found in PPI-treated SW480 cells. Increased levels of ROS might link to autophagy and apoptosis because reducing the level of ROS by antioxidant N-acetylcysteine (NAC) treatment mitigated PPI-induced autophagy and apoptosis. Although we did not know the molecular mechanism of how PPI induced ROS production, this is the first study to show that PPI induces ROS production and down-regulates the AKT/mTOR pathway, which subsequently promotes the autophagic cell death and apoptosis of colon cancer cells. This present study reports PPI as a potential therapeutic agent for colon cancer and reveals its underlying mechanisms of action.


Autophagic Cell Death , Colonic Neoplasms , Apoptosis , Autophagy , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Diosgenin/analogs & derivatives , Humans , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism
17.
Int J Mol Sci ; 23(16)2022 Aug 21.
Article En | MEDLINE | ID: mdl-36012700

Obesity has achieved the appearance of a global epidemic and is a serious cause for concern. The hypothalamus, as the central regulator of energy homeostasis, plays a critical role in regulating food intake and energy expenditure. In this study, we show that TRIM67 in the hypothalamus was responsive to body-energy homeostasis whilst a deficiency of TRIM67 exacerbated metabolic disorders in high-fat-diet-induced obese mice. We found exacerbated neuroinflammation and apoptosis in the hypothalamus of obese TRIM67 KO mice. We also found reduced BDNF in the hypothalamus, which affected the fat sympathetic nervous system innervation and contributed to lipid accumulation in adipose tissue under high-fat-diet exposure. In this study, we reveal potential implications between TRIM67 and the hypothalamic function responding to energy overuptake as well as a consideration for the therapeutic diagnosis of obesity.


Hypothalamus , Obesity , Tripartite Motif Proteins , Adipose Tissue/metabolism , Animals , Cytoskeletal Proteins/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/metabolism , Tripartite Motif Proteins/genetics
18.
Sci Rep ; 12(1): 11838, 2022 07 12.
Article En | MEDLINE | ID: mdl-35821510

While clinical observations have confirmed a link between the development of neurodegenerative diseases and traumatic brain injuries (TBI), there are currently no treatments available and the underlying mechanisms remain elusive. In response, we have developed an in vitro pendulum trauma model capable of imparting rapid acceleration injuries to neuronal networks grown on microelectrode arrays within a clinically relevant range of g forces, with real-time electrophysiological and morphological monitoring. By coupling a primary physical insult with the quantification of post-impact levels of known biochemical pathological markers, we demonstrate the capability of our system to delineate and investigate the primary and secondary injury mechanisms leading to post-impact neurodegeneration. Specifically, impact experiments reveal significant, force-dependent increases in the pro-inflammatory, oxidative stress marker acrolein at 24 h post-impact. The elevation of acrolein was augmented by escalating g force exposures (30-200 g), increasing the number of rapidly repeated impacts (4-6 s interval, 3, 5 and 10×), and by exposing impacted cells to 40 mM ethanol, a known comorbidity of TBI. The elevated levels of acrolein following multiple impacts could be reduced by increasing time-intervals between repeated hits. In addition, we show that conditioned media from maximally-impacted cultures can cause cellular acrolein elevation when introduced to non-impact, control networks, further solidifying acrolein's role as a diffusive-factor in post-TBI secondary injuries. Finally, morphological data reveals post-impact acrolein generation to be primarily confined to soma, with some emergence in cellular processes. In conclusion, this novel technology provides accurate, physical insults with a unique level of structural and temporal resolution, facilitating the investigation of post-TBI neurodegeneration.


Acrolein , Brain Injuries, Traumatic , Acceleration , Humans , Lab-On-A-Chip Devices , Physical Examination
19.
Free Radic Biol Med ; 184: 66-73, 2022 05 01.
Article En | MEDLINE | ID: mdl-35398493

Oxidative stress has been shown to play a critical pathogenic role in functional loss after spinal cord injury (SCI). As a direct result of oxidative stress, lipid peroxidation-derived aldehydes have emerged as key culprits that sustain secondary injury and contribute significantly to pathological outcomes. Acrolein, a neurotoxin, has been shown to be elevated in SCI and can result in post-SCI neurological deficits. Reducing acrolein has therefore emerged as a novel and effective therapeutic strategy in SCI. Previous studies have revealed that hydralazine, an FDA approved blood pressure lowering medication, when administered after SCI shows strong acrolein scavenging capabilities and significantly improves cellular and behavioral outcomes. However, while effective at scavenging acrolein, hydralazine's blood pressure lowering activity can have a detrimental impact on neurotrauma patients. Here, our goal was to preserve the acrolein scavenging capability while mitigating the effect of hydralazine on blood pressure. We accomplished this using a folate-targeted delivery system to deploy hydralazine to the folate receptor positive inflammatory site of the cord injury. Using a model of rat SCI, we found that this system is effective for targeting the injury site, and that folate targeted hydralazine can scavenge acrolein without significantly impacting blood pressure.


Acrolein , Spinal Cord Injuries , Animals , Folic Acid , Humans , Hydralazine/pharmacology , Hydralazine/therapeutic use , Lipid Peroxidation/physiology , Rats , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology
20.
Neurobiol Dis ; 168: 105690, 2022 06 15.
Article En | MEDLINE | ID: mdl-35301122

Autism spectrum disorder (ASD) affects ~2% of the population in the US, and monogenic forms of ASD often result in the most severe manifestation of the disorder. Recently, SCN2A has emerged as a leading gene associated with ASD, of which abnormal sleep pattern is a common comorbidity. SCN2A encodes the voltage-gated sodium channel NaV1.2. Predominantly expressed in the brain, NaV1.2 mediates the action potential firing of neurons. Clinical studies found that a large portion of children with SCN2A deficiency have sleep disorders, which severely impact the quality of life of affected individuals and their caregivers. The underlying mechanism of sleep disturbances related to NaV1.2 deficiency, however, is not known. Using a gene-trap Scn2a-deficient mouse model (Scn2atrap), we found that Scn2a deficiency results in increased wakefulness and reduced non-rapid-eye-movement (NREM) sleep. Brain region-specific Scn2a deficiency in the suprachiasmatic nucleus (SCN) containing region, which is involved in circadian rhythms, partially recapitulates the sleep disturbance phenotypes. At the cellular level, we found that Scn2a deficiency disrupted the firing pattern of spontaneously firing neurons in the SCN region. At the molecular level, RNA-sequencing analysis revealed differentially expressed genes in the circadian entrainment pathway including core clock genes Per1 and Per2. Performing a transcriptome-based compound discovery, we identified dexanabinol (HU-211), a putative glutamate receptor modulator, that can partially reverse the sleep disturbance in mice. Overall, our study reveals possible molecular and cellular mechanisms underlying Scn2a deficiency-related sleep disturbances, which may inform the development of potential pharmacogenetic interventions for the affected individuals.


Autism Spectrum Disorder , Autistic Disorder , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Animals , Autism Spectrum Disorder/genetics , Circadian Rhythm , Mice , NAV1.2 Voltage-Gated Sodium Channel/genetics , Quality of Life , Sleep
...