Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.702
Filter
1.
Adv Exp Med Biol ; 1456: 27-48, 2024.
Article in English | MEDLINE | ID: mdl-39261422

ABSTRACT

Major depressive disorder (MDD) is a mental health disorder associated with cognitive impairment, dysregulated appetite, fatigue, insomnia or hypersomnia, and severe mood changes that significantly impact the ability of the affected individual to perform day-to-day tasks, leading to suicide in the worst-case scenario. As MDD is becoming more prevalent, affecting roughly 300 million individuals worldwide, its treatment has become a major point of interest. Antidepressants acting as selective serotonin reuptake inhibitors (SSRIs) are currently used as the first line of treatment for MDD. Other antidepressants currently used for the treatment of MDD include the serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs). However, although effective in alleviating symptoms of MDD, most antidepressants require weeks or even months of regular administration prior to eliciting a rational clinical effect. Owing to the strong evidence showing a relationship between neural plasticity, neurogenesis, and MDD, researchers have also looked at the possibility of using treatment modalities that target these processes in an attempt to improve clinical outcome. The overarching aim of this chapter is to highlight the role of neural plasticity and neurogenesis in the pathophysiology of MDD and discuss the most recently studied treatment strategies that target these processes by presenting supporting evidence from both animal and human studies.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Neurogenesis , Neuronal Plasticity , Humans , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/physiopathology , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Antidepressive Agents/therapeutic use , Antidepressive Agents/pharmacology , Animals , Selective Serotonin Reuptake Inhibitors/therapeutic use , Selective Serotonin Reuptake Inhibitors/pharmacology
2.
Nat Commun ; 15(1): 7611, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39218970

ABSTRACT

The development of functional neurons is a complex orchestration of multiple signaling pathways controlling cell proliferation and differentiation. Because the balance of antioxidants is important for neuronal survival and development, we hypothesized that ferroptosis must be suppressed to gain neurons. We find that removal of antioxidants diminishes neuronal development and laminar organization of cortical organoids, which is fully restored when ferroptosis is inhibited by ferrostatin-1 or when neuronal differentiation occurs in the presence of vitamin A. Furthermore, iron-overload-induced developmental growth defects in C. elegans are ameliorated by vitamin E and A. We determine that all-trans retinoic acid activates the Retinoic Acid Receptor, which orchestrates the expression of anti-ferroptotic genes. In contrast, retinal and retinol show radical-trapping antioxidant activity. Together, our study reveals an unexpected function of vitamin A in coordinating the expression of essential cellular gatekeepers of ferroptosis, and demonstrates that suppression of ferroptosis by radical-trapping antioxidants or by vitamin A is required to obtain mature neurons and proper laminar organization in cortical organoids.


Subject(s)
Antioxidants , Caenorhabditis elegans , Ferroptosis , Neurons , Vitamin A , Animals , Ferroptosis/drug effects , Vitamin A/pharmacology , Vitamin A/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/drug effects , Antioxidants/pharmacology , Neurons/metabolism , Neurons/drug effects , Neurons/cytology , Cyclohexylamines/pharmacology , Cell Differentiation/drug effects , Vitamin E/pharmacology , Receptors, Retinoic Acid/metabolism , Receptors, Retinoic Acid/genetics , Tretinoin/pharmacology , Organoids/drug effects , Organoids/metabolism , Neurogenesis/drug effects , Mice , Humans , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Signal Transduction/drug effects , Phenylenediamines
3.
Stem Cell Res Ther ; 15(1): 335, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334506

ABSTRACT

Patients with spinal cord injury (SCI) have permanent devastating motor and sensory disabilities. Secondary SCI is known for its complex progression and presents with sophisticated aberrant inflammation, vascular changes, and secondary cellular dysfunction, which aggravate the primary damage. Since their initial discovery, the potent neuroprotective effects and powerful delivery abilities of exosomes (Exos) have been reported in different research fields, including SCI. In this study, we summarize therapeutic advances related to the application of Exos in preclinical animal studies. Subsequently, we discuss the mechanisms of action of Exos derived from diverse cell types, including neurogenesis, angiogenesis, blood-spinal cord barrier preservation, anti-apoptosis, and anti-inflammatory potential. We also evaluate the relationship between the Exo delivery cargo and signaling pathways. Finally, we discuss the challenges and advantages of using Exos to offer innovative insights regarding the development of efficient clinical strategies for SCI.


Subject(s)
Exosomes , Spinal Cord Injuries , Spinal Cord Injuries/therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Exosomes/metabolism , Humans , Animals , Neurogenesis/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology
4.
ACS Chem Neurosci ; 15(19): 3449-3458, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39265183

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) have emerged as a promising target for the treatment of various neurodegenerative disorders. Studies have shown that both PPAR α & γ individually modulate various pathophysiological events like neuroinflammation and insulin resistance, which are known to variedly affect neurogenesis. Our study aimed to evaluate the effect of saroglitazar (SGZR), a dual PPAR agonist, on adult neurogenesis and spatial learning and memory, in intracerebroventricular streptozotocin (ICV STZ)-induced dementia in rats. We have found that SGZR at the dose of 4 mg/kg per oral showed significant improvement in learning and memory compared to ICV STZ-treated rats. A substantial increase in neurogenesis was observed in the subventricular zone (SVZ) and the dentate gyrus (DG), as indicated by an increase in the number of 5-bromo-2'-deoxyuridine (BrdU)+ cells, BrdU+ nestin+ cells, and doublecortin (DCX)+cells. Treatment with SGZR also decreased the active form of glycogen synthase kinase 3ß (GSK3ß) and hence enhanced the nuclear translocation of the ß-catenin. Enhanced expression of Wnt transcription factors and target genes indicates that the up-regulation of Wnt signaling might be involved in the observed increase in neurogenesis. Hence, it can be concluded that the SGZR enhances memory functions and adult neurogenesis via the upregulation of Wnt ß-catenin signaling in ICV STZ-treated rats.


Subject(s)
Dementia , Doublecortin Protein , Memory , Neurogenesis , Up-Regulation , Wnt Signaling Pathway , Animals , Neurogenesis/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , Rats , Male , Up-Regulation/drug effects , Memory/drug effects , Dementia/metabolism , Dementia/chemically induced , Disease Models, Animal , Phenylpropionates/pharmacology , beta Catenin/metabolism , Rats, Wistar , Streptozocin
5.
ACS Chem Neurosci ; 15(19): 3482-3495, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39288278

ABSTRACT

Aging and various neurodegenerative diseases cause significant reduction in adult neurogenesis and simultaneous increase in quiescent neural stem cells (NSCs), which impact the brain's regenerative capabilities. To deal with this challenging issue, current treatments involve stem cell transplants or prevention of neurodegeneration; however, the efficacy or success of this process remains limited. Therefore, extensive and focused investigation is highly demanding to overcome this challenging task. Here, we have designed an efficient peptide-based EphA4 receptor-targeted ligand through an in silico approach. Further, this strategy involves chemical conjugation of the peptide with adipose tissue stem cell-derived EV (Exo-pep-11). Interestingly, our newly designed engineered EV, Exo-pep-11, targets NSC through EphA4 receptors, which offers promising therapeutic advantages by stimulating NSC proliferation and subsequent differentiation. Our result demonstrates that NSC successfully internalized Exo-pep-11 in both in vitro culture conditions as well as in the in vivo aging rats. We found that the uptake of Exo-pep-11 decreased by ∼2.3-fold when NSC was treated with EphA4 antibody before Exo-pep-11 incubation, which confirms the receptor-specific uptake of Exo-pep-11. Exo-pep-11 treatment also increases NSC proliferation by ∼1.9-fold and also shows ∼1.6- and ∼2.4-fold increase in expressions of Nestin and ID1, respectively. Exo-pep-11 also has the potential to increase neurogenesis in aging rats, which is confirmed by ∼1.6- and ∼1.5-fold increases in expressions of TH and Tuj1, respectively, in rat olfactory bulb. Overall, our findings highlight the potential role of Exo-pep-11 for prospective applications in combating age-related declines in NSC activity and neurogenesis.


Subject(s)
Aging , Extracellular Vesicles , Neural Stem Cells , Neurogenesis , Receptor, EphA4 , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Rats , Aging/drug effects , Receptor, EphA4/metabolism , Neurogenesis/physiology , Neurogenesis/drug effects , Extracellular Vesicles/metabolism , Adult Stem Cells/drug effects , Peptides/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Rejuvenation/physiology , Rats, Sprague-Dawley
6.
J Nanobiotechnology ; 22(1): 564, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39272097

ABSTRACT

Intracerebral hemorrhage is a lethal cerebrovascular disease, and the inevitable secondary brain injury (SBI) is responsible for serious disability and death. Perfect therapeutic goal is to minimize SBI and restore neurobehavioral functions. Recently, neuroprotection is highlighted to reduce SBI, but it still faces "Neuronal survival but impaired functions" dilemma. Herein, this work further proposes a novel combinational therapeutic strategy of neuroprotection and neurogenesis toward this goal. However, appropriate therapeutic agents are rarely reported, and their discovery and development are urgently needed. Selenium participates in various physiological/pathological processes, which is hypothesized as a potential targeting molecule. To explore this effect, this work formulates an ultra-small selenium nanodot with a seleno-amino acid derived carbon dot domain and a hydrophilic PEG layer, surprisingly finding that it increases various selenoproteins levels at perihematomal region, to not only exert multiple neuroprotective roles at acute phase but promote neurogenesis and inhibit glial scar formation at recovery phase. At a safe dose, this combinational strategy effectively prevents SBI and recovers neurobehavioral functions to a normal level. Furthermore, its molecular mechanisms are revealed to broaden application scopes in other complex diseases.


Subject(s)
Brain Injuries , Hemorrhagic Stroke , Neuroprotective Agents , Selenium , Animals , Selenium/chemistry , Selenium/pharmacology , Selenium/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Brain Injuries/drug therapy , Hemorrhagic Stroke/drug therapy , Neurogenesis/drug effects , Male , Mice , Selenoproteins/metabolism , Nanoparticles/chemistry , Neurons/drug effects , Brain/drug effects
7.
Ecotoxicol Environ Saf ; 284: 116876, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39146594

ABSTRACT

The prevalence of environmental problems and the increasing risk of human exposure to environmental pollutants have become a global concern. The increasing environmental pollution is one of the main reasons for the rising incidence of most neurological-related diseases in recent years. However, the ethical constraints of direct human research and the racial limitations of animal models have slowed the progress of research in this area. The purpose of this study is to review the neurotoxicity of different environmental pollutants on the brain using brain organoids as a new model and to conclude that brain organoids may play a key role in assessing the mechanisms by which environmental pollutants affect neurogenesis and cause neurological pathogenesis. To accurately determine the negative effects of environmental pollutants on the nervous system, self-organizing brain organoids that are highly similar to the developing brain have become a new model system for studying the effects of environmental pollutants on human brain development and disease. This study uses brain organoids as a model to summarize the neurotoxicity of different environmental pollutants on the nervous system, including structural changes in brain organoids, inhibition of neuronal differentiation and migration, impairment of mitochondrial function, damage to cellular cilia, and influence on signaling pathways. In conclusion, exposure to environmental pollutants may cause different neurotoxicity to the nervous system. Therefore, it is crucial to understand how to use brain organoids to ameliorate neurological disorders caused by environmental pollution.


Subject(s)
Brain , Environmental Pollutants , Organoids , Organoids/drug effects , Environmental Pollutants/toxicity , Humans , Brain/drug effects , Animals , Neurogenesis/drug effects , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/etiology , Models, Biological
8.
Exp Neurol ; 381: 114930, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39173898

ABSTRACT

Traumatic brain injury (TBI) presents a significant public health challenge, necessitating innovative interventions for effective treatment. Recent studies have challenged conventional perspectives on neurogenesis, unveiling endogenous repair mechanisms within the adult brain following injury. However, the intricate mechanisms governing post-TBI neurogenesis remain unclear. The microenvironment of an injured brain, characterized by astrogliosis, neuroinflammation, and excessive cell death, significantly influences the fate of newly generated neurons. Adenosine kinase (ADK), the key metabolic regulator of adenosine, emerges as a crucial factor in brain development and cell proliferation after TBI. This study investigates the hypothesis that targeting ADK could enhance brain repair, promote neuronal survival, and facilitate differentiation. In a TBI model induced by controlled cortical impact, C57BL/6 male mice received intraperitoneal injections of the small molecule ADK inhibitor 5-iodotubercidin (ITU) for three days following TBI. To trace the fate of TBI-associated proliferative cells, animals received intraperitoneal injections of BrdU for seven days, beginning immediately after TBI. Our results show that ADK inhibition by ITU improved brain repair 14 days after injury as evidenced by a diminished injury size. Additionally, the number of mature neurons generated after TBI was increased in ITU-treated mice. Remarkably, the TBI-associated pathological events including astrogliosis, neuroinflammation, and cell death were arrested in ITU-treated mice. Finally, ADK inhibition modulated cell death by regulating the PERK signaling pathway. Together, these findings demonstrate a novel therapeutic approach to target multiple pathological mechanisms involved in TBI. This research contributes valuable insights into the intricate molecular mechanisms underlying neurogenesis and gliosis after TBT.


Subject(s)
Adenosine Kinase , Brain Injuries, Traumatic , Cell Differentiation , Cell Survival , Mice, Inbred C57BL , Neurogenesis , Neurons , Animals , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Male , Mice , Adenosine Kinase/antagonists & inhibitors , Adenosine Kinase/metabolism , Cell Differentiation/drug effects , Neurons/drug effects , Neurons/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Tubercidin/analogs & derivatives
9.
Mil Med Res ; 11(1): 61, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39169440

ABSTRACT

BACKGROUND: Chronic Gulf War Illness (GWI) is characterized by cognitive and mood impairments, as well as persistent neuroinflammation and oxidative stress. This study aimed to investigate the efficacy of Epidiolex®, a Food and Drug Administration (FDA)-approved cannabidiol (CBD), in improving brain function in a rat model of chronic GWI. METHODS: Six months after exposure to low doses of GWI-related chemicals [pyridostigmine bromide, N,N-diethyl-meta-toluamide (DEET), and permethrin (PER)] along with moderate stress, rats with chronic GWI were administered either vehicle (VEH) or CBD (20 mg/kg, oral) for 16 weeks. Neurobehavioral tests were conducted on 11 weeks after treatment initiation to evaluate the performance of rats in tasks related to associative recognition memory, object location memory, pattern separation, and sucrose preference. The effect of CBD on hyperalgesia was also examined. The brain tissues were processed for immunohistochemical and molecular studies following behavioral tests. RESULTS: GWI rats treated with VEH exhibited impairments in all cognitive tasks and anhedonia, whereas CBD-treated GWI rats showed improvements in all cognitive tasks and no anhedonia. Additionally, CBD treatment alleviated hyperalgesia in GWI rats. Analysis of hippocampal tissues from VEH-treated rats revealed astrocyte hypertrophy and increased percentages of activated microglia presenting NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) complexes as well as elevated levels of proteins involved in NLRP3 inflammasome activation and Janus kinase/signal transducers and activators of the transcription (JAK/STAT) signaling. Furthermore, there were increased concentrations of proinflammatory and oxidative stress markers along with decreased neurogenesis. In contrast, the hippocampus from CBD-treated GWI rats displayed reduced levels of proteins mediating the activation of NLRP3 inflammasomes and JAK/STAT signaling, normalized concentrations of proinflammatory cytokines and oxidative stress markers, and improved neurogenesis. Notably, CBD treatment did not alter the concentration of endogenous cannabinoid anandamide in the hippocampus. CONCLUSIONS: The use of an FDA-approved CBD (Epidiolex®) has been shown to effectively alleviate cognitive and mood impairments as well as hyperalgesia associated with chronic GWI. Importantly, the improvements observed in rats with chronic GWI in this study were attributed to the ability of CBD to significantly suppress signaling pathways that perpetuate chronic neuroinflammation.


Subject(s)
Cannabidiol , Cognitive Dysfunction , Hyperalgesia , Neurogenesis , Neuroinflammatory Diseases , Persian Gulf Syndrome , Animals , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Rats , Persian Gulf Syndrome/drug therapy , Persian Gulf Syndrome/complications , Male , Hyperalgesia/drug therapy , Neuroinflammatory Diseases/drug therapy , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Neurogenesis/drug effects , Disease Models, Animal , Rats, Sprague-Dawley , Signal Transduction/drug effects , Mood Disorders/drug therapy , Oxidative Stress/drug effects , Hippocampus/drug effects , Pyridostigmine Bromide/pharmacology , Pyridostigmine Bromide/therapeutic use
10.
Eur J Med Chem ; 277: 116763, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39146834

ABSTRACT

5-Methoxy-3-(5-methoxyindolin-2-yl)-1H-indole (3), whose structure was unambiguously elucidated by X-ray analysis, was identified as a multi-target compound with potential application in neurodegenerative diseases. It is a low nanomolar inhibitor of QR2 (IC50 = 7.7 nM), with greater potency than melatonin and comparable efficacy to the most potent QR2 inhibitors described to date. Molecular docking studies revealed the potential binding mode of 3 to QR2, which explains its superior potency compared to melatonin. Furthermore, compound 3 inhibits hMAO-A, hMAO-B and hLOX-5 in the low micromolar range and is an excellent ROS scavenger. In phenotypic assays, compound 3 showed neuroprotective activity in a cellular model of oxidative stress damage, it was non-toxic, and was able to activate neurogenesis from neural stem-cell niches of adult mice. These excellent biological properties, together with its both good in silico and in vitro drug-like profile, highlight compound 3 as a promising drug candidate for neurodegenerative diseases.


Subject(s)
Melatonin , Molecular Docking Simulation , Neurogenesis , Neuroprotective Agents , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Melatonin/pharmacology , Melatonin/chemistry , Animals , Mice , Humans , Structure-Activity Relationship , Neurogenesis/drug effects , Molecular Structure , Drug Discovery , Quinone Reductases/antagonists & inhibitors , Quinone Reductases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Dose-Response Relationship, Drug
11.
Neuroscience ; 558: 1-10, 2024 Oct 18.
Article in English | MEDLINE | ID: mdl-39137868

ABSTRACT

The Ribosomal S6 Kinase (RSK) family of serine/threonine kinases function as key downstream effectors of the MAPK signaling cascade. In the nervous system, RSK signaling plays crucial roles in neuronal development and contributes to activity-dependent neuronal plasticity. This study examined the role of RSK signaling in cell viability during neuronal development and in neuroprotection in the mature nervous system. Using neuronal cell-culture-based profiling, we found that suppressing RSK signaling led to significant cell death in developing primary neuronal cultures. To this end, treatment with the RSK inhibitors BiD1870 or SL0101 on the first day of culturing resulted in over 80% cell death. In contrast, more mature cultures showed attenuated cell death upon RSK inhibition. Inhibition of RSK signaling during early neuronal development also disrupted neurite outgrowth and cell growth. In maturing hippocampal explant cultures, treatment with BiD1870 had minimal effects on cell viability, but led to a striking augmentation of NMDA-induced cell death. Finally, we used the endothelin 1 (ET-1) model of ischemia to examine the neuroprotective effects of RSK signaling in the mature hippocampus in vivo. Notably, in the absence of RSK inhibition, the granule cell layer (GCL) was resistant to the effects of ET-1; However, disruption of RSK signaling (via the microinjection of BiD1870) prior to ET-1 injection triggered cell death within the GCL, thus indicating a neuroprotective role for RSK signaling in the mature nervous system. Together these data reveal distinct, developmentally-defined, roles for RSK signaling in the nervous system.


Subject(s)
Cell Death , Cell Survival , Neurons , Ribosomal Protein S6 Kinases , Signal Transduction , Animals , Neurons/drug effects , Neurons/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Cells, Cultured , Endothelin-1/pharmacology , Endothelin-1/metabolism , N-Methylaspartate/pharmacology , Rats, Sprague-Dawley , Rats , Neurogenesis/physiology , Neurogenesis/drug effects , Pteridines
12.
Ecotoxicol Environ Saf ; 283: 116831, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39151374

ABSTRACT

BACKGROUND: Aflatoxin B1, which can penetrate the blood-brain barrier and kill neural cells, can contaminate traditional herbal medicines, posing a significant risk to human health. The present study examined cellular, cognitive and behavioral consequences of aflatoxin B1 contamination of the anti-osteoporotic medicine Radix Dipsaci. METHODS: A mouse model of osteoporosis was created by treating the animals with all-trans-retinoic acid. Then the animals were treated intragastically with water decoctions of Radix Dipsaci that contained detectable aflatoxin B1 or not. The animals were compared in terms of mineral density and mineral salt content of bone, production of pro-inflammatory factors, neurogenesis and microglial activation in hippocampus, as well as behavior and cognitive function. RESULTS: Contamination of Radix Dipsaci with aflatoxin B1 significantly reduced the medicine's content of bioactive saponins. It destroyed the ability of the herbal decoction to improve mineral density and mineral salt content in the bones of diseased mice, and it induced the production of the oxidative stress marker malondialdehyde as well as the pro-inflammatory cytokines interleukin-1ß and tumor necrosis factor-α. Aflatoxin B1 contamination inhibited formation of new neurons and increased the proportion of activated microglia in the hippocampus. These neurological changes were associated with anhedonia, behavioral despair, and deficits in short-term memory and social memory. CONCLUSION: Contamination of Radix Dipsaci with aflatoxin B1 not only eliminates the herbal decoction's anti-osteoporotic effects, but it also induces neurotoxicity that can lead to cognitive decline and behavioral abnormalities. Such contamination should be avoided through tightly regulated production and quality control of medicinal herbs.


Subject(s)
Aflatoxin B1 , Cognition , Disease Models, Animal , Hippocampus , Neurogenesis , Osteoporosis , Animals , Hippocampus/drug effects , Aflatoxin B1/toxicity , Mice , Osteoporosis/drug therapy , Osteoporosis/chemically induced , Cognition/drug effects , Neurogenesis/drug effects , Dipsacaceae/chemistry , Male , Drug Contamination , Drugs, Chinese Herbal/pharmacology
13.
Int J Neuropsychopharmacol ; 27(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39185814

ABSTRACT

BACKGROUND: Depression is a heterogeneous disorder with high morbidity and disability rates that poses serious problems regarding mental health care. It is now well established that N-methyl D-aspartate receptor (NMDAR) modulators are being increasingly explored as potential therapeutic options for treating depression, although relatively little is known about their mechanisms of action. NMDARs are glutamate-gated ion channels that are ubiquitously expressed in the central nervous system (CNS), and they have been shown to play key roles in excitatory synaptic transmission. GluN2A, the predominant Glu2N subunit of functional NMDARs in neurons, is involved in various physiological processes in the CNS and is associated with diseases such as anxiety, depression, and schizophrenia. However, the role of GluN2A in the pathophysiology of depression has not yet been elucidated. METHODS: We reviewed several past studies to better understand the function of GluN2A in depression. Additionally, we also summarized the pathogenesis of depression based on the regulation of GluN2A expression, particularly its interaction with neuroinflammation and neurogenesis, which has received considerable critical attention and is highly implicated in the onset of depression. RESULTS: These evidence suggests that GluN2A overexpression impairs structural and functional synaptic plasticity, which contributes to the development of depression. Consequently, this knowledge is vital for the development of selective antagonists targeting GluN2A subunits using pharmacological and molecular methods. CONCLUSIONS: Specific inhibition of the GluN2A NMDAR subunit is resistant to chronic stress-induced depressive-like behaviors, making them promising targets for the development of novel antidepressants.


Subject(s)
Antidepressive Agents , Receptors, N-Methyl-D-Aspartate , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Humans , Antidepressive Agents/pharmacology , Animals , Depression/drug therapy , Depression/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Drug Development , Neurogenesis/drug effects
14.
BMC Complement Med Ther ; 24(1): 293, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090706

ABSTRACT

BACKGROUND: Salidroside is the major bioactive and pharmacological active substance in Rhodiola rosea L. It has been reported to have neuroprotective effects on cerebral ischemia/reperfusion (I/R). However, whether salidroside can enhance neural regeneration after cerebral I/R is still unknown. This study investigated the effects of salidroside on the endogenous neural regeneration after cerebral I/R and the related mechanism. METHODS: Focal cerebral I/R was induced in rats by transient middle cerebral artery occlusion/reperfusion (MCAO/R). The rats were intraperitoneally treated salidroside once daily for 7 consecutive days. Neurobehavioral assessments were performed at 3 days and 7 days after the injury. TTC staining was performed to assess cerebral infarct volume. To evaluate the survival of neurons, immunohistochemical staining of Neuronal Nuclei (NeuN) in the ischemic hemisphere were conducted. Also, immunofluorescence double or triple staining of the biomarkers of proliferating neural progenitor cells in Subventricular Zone (SVZ) and striatum of the ischemia hemisphere were performed to investigate the neurogenesis. Furthermore, reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression of neurotrophic factors (NTFs) brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). Expression of Notch1 and its target molecular Hes1 were also analyzed by western-blotting and RT-PCR. RESULTS: Salidroside treatment ameliorated I/R induced neurobehavioral impairment, and reduced infarct volume. Salidroside also restored NeuN positive cells loss after I/R injury. Cerebral I/R injury significantly increased the expression of 5-Bromo-2'-Deoxyuridine (BrdU) and doublecotin (DCX), elevated the number of BrdU/Nestin/DCX triple-labeled cells in SVZ, and BrdU/Nestin/glial fibrillary acidic protein (GFAP) triple-labeled cells in striatum. Salidroside treatment further promoted the proliferation of BrdU/DCX labeled neuroblasts and BrdU/Nestin/GFAP labeled reactive astrocytes. Furthermore, salidroside elevated the mRNA expression and protein concentration of BDNF and NGF in ischemia periphery area, as well. Mechanistically, salidroside elevated Notch1/Hes1 mRNA expression in SVZ. The protein levels of them were also increased after salidroside administration. CONCLUSIONS: Salidroside enhances the endogenous neural regeneration after cerebral I/R. The mechanism of the effect may involve the regulation of BDNF/NGF and Notch signaling pathway.


Subject(s)
Brain Ischemia , Glucosides , Nerve Regeneration , Phenols , Rats, Sprague-Dawley , Reperfusion Injury , Signal Transduction , Animals , Glucosides/pharmacology , Phenols/pharmacology , Rats , Male , Signal Transduction/drug effects , Reperfusion Injury/drug therapy , Brain Ischemia/drug therapy , Nerve Regeneration/drug effects , Neuroprotective Agents/pharmacology , Nerve Growth Factors/metabolism , Disease Models, Animal , Receptors, Notch/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Neurogenesis/drug effects
15.
Cells ; 13(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39195239

ABSTRACT

Autism spectrum disorder (ASD) is a multifactorial neurodevelopmental condition with several identified risk factors, both genetic and non-genetic. Among these, prenatal exposure to valproic acid (VPA) has been extensively associated with the development of the disorder. The zebrafish, a cost- and time-effective model, is useful for studying ASD features. Using validated VPA-induced ASD zebrafish models, we aimed to provide new insights into VPA exposure effects during embryonic development and to identify new potential biomarkers associated with ASD-like features. Dose-response analyses were performed in vivo to study larval phenotypes and mechanisms underlying neuroinflammation, mitochondrial dysfunction, oxidative stress, microglial cell status, and motor behaviour. Wild-type and transgenic Tg(mpeg1:EGFP) zebrafish were water-exposed to VPA doses (5 to 500 µM) from 6 to 120 h post-fertilisation (hpf). Embryos and larvae were monitored daily to assess survival and hatching rates, and numerous analyses and tests were conducted from 24 to 120 hpf. VPA doses higher than 50 µM worsened survival and hatching rates, while doses of 25 µM or more altered morphology, microglial status, and larval behaviours. VPA 50 µM also affected mRNA expression of inflammatory cytokines and neurogenesis-related genes, mitochondrial respiration, and reactive oxygen species accumulation. The study confirmed that VPA alters brain homeostasis, synaptic interconnections, and neurogenesis-related signalling pathways, contributing to ASD aetiopathogenesis. Further studies are essential to identify novel ASD biomarkers for developing new drug targets and tailored therapeutic interventions for ASD.


Subject(s)
Autism Spectrum Disorder , Disease Models, Animal , Valproic Acid , Zebrafish , Animals , Valproic Acid/pharmacology , Valproic Acid/adverse effects , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Larva/drug effects , Animals, Genetically Modified , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Microglia/drug effects , Microglia/pathology , Microglia/metabolism , Brain/drug effects , Brain/pathology , Brain/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Neurogenesis/drug effects
16.
Nutrients ; 16(16)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39203863

ABSTRACT

Benzo[a]pyrene (B[a]P) is known to inhibit neurodifferentiation and induce neurodegeneration. Agarwood or Aquilaria crassna (AC), a plant with health-promoting properties, may counteract the neurotoxic effects of B[a]P by promoting neuronal growth and survival. This study investigated the protective effect of AC leaf ethanolic extract (ACEE) on the B[a]P-induced impairment of neuronal differentiation. A transcriptomic analysis identified the canonical pathway, the biological network, and the differentially expressed genes (DEGs) that are changed in response to neuronal differentiation and neurogenesis. Several genes, including CXCR4, ENPP2, GAP43, GFRA2, NELL2, NFASC, NSG2, NGB, BASP1, and NEUROD1, in B[a]P-treated SH-SY5Y cells were up-regulated after treatment with ACEE. Notably, a Western blot analysis further confirmed that ACEE increased the protein levels of GAP43 and neuroglobin. B[a]P treatment led to decreased phosphorylation of Akt and increased phosphorylation of ERK in SH-SY5Y cells; however, ACEE was able to reverse these effects. Clionasterol and lupenone were identified in ACEE. Molecular docking showed that these two phytochemicals had significant interactions with CXCR4, GDNF family receptor alpha (GFRA), and retinoid X receptors (RXRs). In conclusion, ACEE may be a potential alternative medicine for the prevention of impaired neuronal differentiation and neurodegenerative diseases.


Subject(s)
Benzo(a)pyrene , Neuroprotective Agents , Plant Extracts , Thymelaeaceae , Humans , Plant Extracts/pharmacology , Neuroprotective Agents/pharmacology , Benzo(a)pyrene/toxicity , Cell Line, Tumor , Thymelaeaceae/chemistry , Gene Expression Profiling , RNA-Seq , Plant Leaves/chemistry , Neurons/drug effects , Neurons/metabolism , Transcriptome/drug effects , Cell Differentiation/drug effects , Neurogenesis/drug effects , Molecular Docking Simulation , Cell Survival/drug effects
17.
Sci Rep ; 14(1): 19347, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164321

ABSTRACT

The aim of this study was to investigate the possible protective effects of apelin, which is known to have antioxidant and anti-inflammatory effects, on changes in neurogenesis in newborns of pregnant rats with L-NAME-induced preeclampsia. Wistar albino female rats were divided into four experimental groups: Control, Apelin, Preeclampsia and Preeclampsia + Apelin. Blood pressure was measured on the 5th, 11th and 17th days of gestation, urine protein was analyzed from urine samples collected for 24 h on the 6th, 12th and 18th days and serum creatinine was analyzed from serum samples. Maternal kidney and placenta tissues were obtained to establish the preeclampsia model, and neonatal brain tissues including the cortex, hippocampus and cerebellum regions were obtained to investigate neurogenesis and examined by histological and immunohistochemical methods. The number of newborns, body weight and brain weight of the newborns were measured. eNOS, IL-10, nNOS and NO levels in the brain analyzed via ELISA. Mean arterial pressure, urine protein and serum creatinine increased in the preeclampsia. Newborn weight decreased in the Preeclampsia group, the values in the Preeclampsia + Apelin group were closer to the Control and Apelin groups. In the Preeclampsia group, edema and dilatation in the proximal and distal tubules of kidneys, perivillous fibrin deposition and increase in syncytial nodules of placenta were observed. VEGF immunoreactivity decreased and iNOS immunoreactivity increased in both kidney and placenta. In neonatal brain tissue examinations, cytotoxic edema accompanied by thinning of cortex, delayed migration and lower cell counts in the hippocampus, and increase in intercellular spaces and EGL thickening in the cerebellum were observed in the preeclampsia. Expression of NeuN, GFAP, MBP, IL-10, eNOS, nNOS and NO levels decreased, whereas expression of Iba-1 increased in the preeclampsia. In the Preeclampsia + Apelin group, these findings were similar to the Control and Apelin groups. Apelin administration was found to be beneficial for preventing the adverse consequences of preeclampsia, but further experimental and clinical studies are needed to better understand these effects.


Subject(s)
Animals, Newborn , Apelin , Brain , NG-Nitroarginine Methyl Ester , Neurogenesis , Pre-Eclampsia , Rats, Wistar , Female , Pregnancy , Pre-Eclampsia/chemically induced , Pre-Eclampsia/metabolism , Animals , Apelin/metabolism , Neurogenesis/drug effects , Rats , Brain/metabolism , Brain/pathology , Brain/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Placenta/metabolism , Disease Models, Animal , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Interleukin-10/metabolism , Interleukin-10/blood , Nitric Oxide Synthase Type I/metabolism
18.
Neuropharmacology ; 258: 110082, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39009217

ABSTRACT

Exercise is known to reduce depression and anxiety symptoms. Although the cellular and molecular mechanisms underlying this effect remain unknown, exercise-induced increases in neurotransmitter release and hippocampal neurogenesis have been hypothesized to play key roles. One neurotransmitter that has been implicated in both antidepressant-like effects and the regulation of hippocampal neurogenesis is serotonin (5-HT). Complete loss of function of the brain 5-HT synthesis enzyme (tryptophan hydroxylase 2, Tph2) has been reported to prevent exercise-induced increases in neurogenesis and to block a subset of antidepressant-like responses to selective serotonin reuptake inhibitors (SSRIs), but whether partial loss of Tph2 function blocks the behavioral and neurogenic effects of exercise has not been established. This study used four tests that are predictive of antidepressant efficacy to determine the impact of 5-HT deficiency on responses to exercise in male and female mice. Our results demonstrate that low 5-HT impairs the behavioral effects of exercise in females in the forced swim and novelty-suppressed feeding tests. However, genetic reductions in 5-HT synthesis did not significantly impact exercise-induced alterations in cellular proliferation or immature neuron production in the hippocampus in either sex. These findings highlight the importance of brain 5-HT in mediating behavioral responses to exercise and suggest that individual differences in brain 5-HT synthesis could influence sensitivity to the mental health benefits of exercise. Furthermore, the observed disconnect between neurogenic and behavioral responses to exercise suggests that increased neurogenesis is unlikely to be the primary driver of the behavioral effects of exercise observed here.


Subject(s)
Neurogenesis , Physical Conditioning, Animal , Serotonin , Tryptophan Hydroxylase , Animals , Tryptophan Hydroxylase/metabolism , Tryptophan Hydroxylase/genetics , Neurogenesis/physiology , Neurogenesis/drug effects , Serotonin/metabolism , Male , Female , Physical Conditioning, Animal/physiology , Mice , Hippocampus/metabolism , Hippocampus/drug effects , Mice, Transgenic , Brain/metabolism , Brain/drug effects , Mice, Inbred C57BL , Gene Knock-In Techniques , Behavior, Animal/drug effects , Behavior, Animal/physiology
19.
Food Funct ; 15(16): 8310-8329, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39069830

ABSTRACT

Cocoa is widely known for its health benefits, but its neurocognitive impact remains underexplored. This preclinical study aimed to investigate the effects of cocoa and cocoa polyphenols on hippocampal neuroplasticity, cognitive function and emotional behavior. Seventy young-adult C57BL/6JRj male and female mice were fed either a standard diet (CTR) or a diet enriched with 10% high-phenolic content cocoa (HPC) or low-phenolic content cocoa (LPC) for at least four weeks. In a first experiment, behavioral tests assessing exploratory behavior, emotional responses and hippocampal-dependent memory were conducted four weeks into the diet, followed by animal sacrifice a week later. Adult hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) expression in the hippocampus and prefrontal cortex were evaluated using immunohistochemistry and western blot. In a different experiment, hippocampal synaptic response, long-term potentiation and presynaptic-dependent short-term plasticity were studied by electrophysiology. Cocoa-enriched diets had minimal effects on exploratory activity and anxiety-like behavior, except for reduced locomotion in the LPC group. Only the HPC diet enhanced object recognition memory, while place recognition memory and spatial navigation remained unaffected. The HPC diet also increased adult hippocampal neurogenesis, boosting the proliferation, survival and number of young adult-born neurons. However, both cocoa-enriched diets increased immobility in the forced swimming test and hippocampal BDNF expression. Hippocampal electrophysiology revealed no alterations in neuroplasticity among diets. The results were mostly unaffected by sex. Overall, the HPC diet demonstrated greater potential regarding cognitive and neuroplastic benefits, suggesting a key role of cocoa flavanols in dietary interventions aimed at enhancing brain health.


Subject(s)
Brain-Derived Neurotrophic Factor , Cacao , Hippocampus , Memory , Mice, Inbred C57BL , Neurogenesis , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Neurogenesis/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Mice , Male , Female , Memory/drug effects , Cacao/chemistry , Neuronal Plasticity/drug effects , Diet
20.
Pharmacol Biochem Behav ; 243: 173821, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002805

ABSTRACT

Schizophrenia impacts about 1 % of the global population, with clozapine (CLZ) being a critical treatment for refractory cases despite its limitations in effectiveness and adverse effects. Therefore, the search for more effective treatments remains urgent. Light treatment (LT) recognized for enhancing cognition and mood, presents a promising complementary approach. This study investigated the effects of CLZ and LT on cognitive impairments in a sub-chronic MK-801 induced schizophrenia mouse model. Results showed that both CLZ and CLZ + LT treatment elevate cognitive performance of sub-chronic MK-801 treated mice in serial behavioral tests over two months. Histological analysis revealed increased dendritic spine density and branching, and synaptic repair in the hippocampus with CLZ and CLZ + LT interventions. Furthermore, both treatments increased brain-derived neurotrophic factor (BDNF) expression in the hippocampus, likely contributing to cognitive amelioration in MK-801 treated mice. Additionally, BrdU labeling revealed that CLZ + LT further enhances neurogenesis in the dentate gyrus (DG) and lateral ventricle (LV) of sub-chronic MK-801 treated mice. These findings may have implications for the development of noninvasive and adjunctive treatment strategies aimed at alleviating cognitive impairments and improving functional outcomes in individuals with schizophrenia.


Subject(s)
Antipsychotic Agents , Clozapine , Cognitive Dysfunction , Dizocilpine Maleate , Hippocampus , Neurogenesis , Neuronal Plasticity , Schizophrenia , Animals , Clozapine/pharmacology , Dizocilpine Maleate/pharmacology , Mice , Neurogenesis/drug effects , Male , Cognitive Dysfunction/drug therapy , Schizophrenia/drug therapy , Neuronal Plasticity/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Antipsychotic Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Light , Combined Modality Therapy
SELECTION OF CITATIONS
SEARCH DETAIL