Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.269
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1362573, 2024.
Article in English | MEDLINE | ID: mdl-38645426

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a key neurotrophin within the brain, by selectively activating the TrkB receptor, exerts multimodal effects on neurodevelopment, synaptic plasticity, cellular integrity and neural network dynamics. In parallel, glucocorticoids (GCs), vital steroid hormones, which are secreted by adrenal glands and rapidly diffused across the mammalian body (including the brain), activate two different groups of intracellular receptors, the mineralocorticoid and the glucocorticoid receptors, modulating a wide range of genomic, epigenomic and postgenomic events, also expressed in the neural tissue and implicated in neurodevelopment, synaptic plasticity, cellular homeostasis, cognitive and emotional processing. Recent research evidences indicate that these two major regulatory systems interact at various levels: they share common intracellular downstream pathways, GCs differentially regulate BDNF expression, under certain conditions BDNF antagonises the GC-induced effects on long-term potentiation, neuritic outgrowth and cellular death, while GCs regulate the intraneuronal transportation and the lysosomal degradation of BDNF. Currently, the BDNF-GC crosstalk features have been mainly studied in neurons, although initial findings show that this crosstalk could be equally important for other brain cell types, such as astrocytes. Elucidating the precise neurobiological significance of BDNF-GC interactions in a tempospatial manner, is crucial for understanding the subtleties of brain function and dysfunction, with implications for neurodegenerative and neuroinflammatory diseases, mood disorders and cognitive enhancement strategies.


Subject(s)
Brain-Derived Neurotrophic Factor , Glucocorticoids , Humans , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/physiology , Glucocorticoids/metabolism , Animals , Brain/metabolism , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/metabolism , Signal Transduction , Neurons/metabolism
2.
CNS Neurol Disord Drug Targets ; 23(3): 353-366, 2024.
Article in English | MEDLINE | ID: mdl-37287291

ABSTRACT

The burden of neurological illnesses on global health is significant. Our perception of the molecular and biological mechanisms underlying intellectual processing and behavior has significantly advanced over the last few decades, laying the groundwork for potential therapies for various neurodegenerative diseases. A growing body of literature reveals that most neurodegenerative diseases could be due to the gradual failure of neurons in the brain's neocortex, hippocampus, and various subcortical areas. Research on various experimental models has uncovered several gene components to understand the pathogenesis of neurodegenerative disorders. One among them is the brain-derived neurotrophic factor (BDNF), which performs several vital functions, enhancing synaptic plasticity and assisting in the emergence of long-term thoughts. The pathophysiology of some neurodegenerative diseases, including Alzheimer's, Parkinson's, Schizophrenia, and Huntington's, has been linked to BDNF. According to numerous research, high levels of BDNF are connected to a lower risk of developing a neurodegenerative disease. As a result, we want to concentrate on BDNF in this article and outline its protective role against neurological disorders.


Subject(s)
Brain-Derived Neurotrophic Factor , Neurodegenerative Diseases , Humans , Brain-Derived Neurotrophic Factor/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Protective Agents
3.
PLoS Biol ; 21(4): e3002070, 2023 04.
Article in English | MEDLINE | ID: mdl-37011100

ABSTRACT

During development, patterned neural activity instructs topographic map refinement. Axons with similar patterns of neural activity converge onto target neurons and stabilize their synapses with these postsynaptic partners, restricting exploratory branch elaboration (Hebbian structural plasticity). On the other hand, non-correlated firing in inputs leads to synapse weakening and increased exploratory growth of axons (Stentian structural plasticity). We used visual stimulation to control the correlation structure of neural activity in a few ipsilaterally projecting (ipsi) retinal ganglion cell (RGC) axons with respect to the majority contralateral eye inputs in the optic tectum of albino Xenopus laevis tadpoles. Multiphoton live imaging of ipsi axons, combined with specific targeted disruptions of brain-derived neurotrophic factor (BDNF) signaling, revealed that both presynaptic p75NTR and TrkB are required for Stentian axonal branch addition, whereas presumptive postsynaptic BDNF signaling is necessary for Hebbian axon stabilization. Additionally, we found that BDNF signaling mediates local suppression of branch elimination in response to correlated firing of inputs. Daily in vivo imaging of contralateral RGC axons demonstrated that p75NTR knockdown reduces axon branch elongation and arbor spanning field volume.


Subject(s)
Brain-Derived Neurotrophic Factor , Dendrites , Brain-Derived Neurotrophic Factor/physiology , Dendrites/physiology , Retinal Ganglion Cells/physiology , Axons/physiology , Synapses/physiology
4.
Eur J Med Chem ; 248: 115111, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36645981

ABSTRACT

Numerous studies have been published about the implication of the neurotrophin brain-derived neurotrophic factor (BDNF) and its receptor TrkB in the pathogenesis of several neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, Multiple Sclerosis and motor neuron disease. BDNF activates the TrkB receptor with high potency and specificity, promoting neuronal survival, differentiation and synaptic plasticity. Based on the main structural characteristics of LM22A-4, a previously published small molecule that acts as activator of the TrkB receptor, we have designed and synthesized a small data set of compounds. The lead idea for the design of the new compounds was to modify the third position of the LM22A-4, by introducing different substitutions in order to obtain compounds which will have not only better physicochemical properties but selective activity as well. ADME and toxicity profiles of molecules have been evaluated as well as their biological properties through the TrkB receptor and affinity to promote neurite differentiation.


Subject(s)
Brain-Derived Neurotrophic Factor , Receptor, trkB , Receptor, trkB/metabolism , Brain-Derived Neurotrophic Factor/physiology , Benzamides , Signal Transduction
5.
Proc Natl Acad Sci U S A ; 119(32): e2201151119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35930664

ABSTRACT

Epilepsy is a devastating brain disorder for which effective treatments are very limited. There is growing interest in early intervention, which requires a better mechanistic understanding of the early stages of this disorder. While diverse brain insults can lead to epileptic activity, a common cellular mechanism relies on uncontrolled recurrent excitatory activity. In the dentate gyrus, excitatory mossy cells (MCs) project extensively onto granule cells (GCs) throughout the hippocampus, thus establishing a recurrent MC-GC-MC excitatory loop. MCs are implicated in temporal lobe epilepsy, a common form of epilepsy, but their role during initial seizures (i.e., before the characteristic MC loss that occurs in late stages) is unclear. Here, we show that initial seizures acutely induced with an intraperitoneal kainic acid (KA) injection in adult mice, a well-established model that leads to experimental epilepsy, not only increased MC and GC activity in vivo but also triggered a brain-derived neurotrophic factor (BDNF)-dependent long-term potentiation (LTP) at MC-GC excitatory synapses. Moreover, in vivo induction of MC-GC LTP using MC-selective optogenetic stimulation worsened KA-induced seizures. Conversely, Bdnf genetic removal from GCs, which abolishes LTP, and selective MC silencing were both anticonvulsant. Thus, initial seizures are associated with MC-GC synaptic strengthening, which may promote later epileptic activity. Our findings reveal a potential mechanism of epileptogenesis that may help in developing therapeutic strategies for early intervention.


Subject(s)
Brain-Derived Neurotrophic Factor , Epilepsy , Long-Term Potentiation , Mossy Fibers, Hippocampal , Seizures , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/physiopathology , Kainic Acid/pharmacology , Mice , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/physiopathology , Seizures/chemically induced , Seizures/physiopathology
6.
Sleep Breath ; 26(1): 287-295, 2022 03.
Article in English | MEDLINE | ID: mdl-33993395

ABSTRACT

PURPOSE: Chronic intermittent hypoxia (CIH) plays a key role in the complications of obstructive sleep apnea (OSA), which is strongly associated with retinal and optic nerve diseases. Additionally, the brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling pathway plays an important protective role in neuronal injury. In the present study, we investigated the role of 7,8-dihydroxyflavone (7,8-DHF) in regulating CIH-induced injury in mice retinas and rat primary retinal ganglion cells (RGCs). METHODS: C57BL/6 mice and in vitro primary RGCs were exposed to CIH or normoxia and treated with or without 7,8-DHF. The mice eyeballs or cultured cells were then taken for histochemistry, immunofluorescence or biochemistry, and the protein expression of the BDNF/TrkB signaling pathway analysis. RESULTS: Our results showed that CIH induced oxidative stress (OS) in in vivo and in vitro models and inhibited the conversion of BDNF precursor (pro-BDNF) to a mature form of BDNF, which increased neuronal cell apoptosis. 7,8-DHF reduced the production of reactive oxygen species (ROS) caused by CIH and effectively activated TrkB signals and downstream protein kinase B (Akt) and extracellular signal-regulated kinase (Erk) survival signaling pathways, which upregulated the expression of mature BDNF. ANA-12 (a TrkB specific inhibitor) blocked the protective effect of 7,8-DHF. CONCLUSION: In short, the activation of the BDNF/TrkB signaling pathway alleviated CIH-induced oxidative stress damage of the optic nerve and retinal ganglion cells. 7,8-DHF may serve as a promising agent for OSA related neuropathy.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/physiology , Cell Hypoxia/drug effects , Flavones/pharmacology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Receptor, trkB/drug effects , Receptor, trkB/physiology , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , Animals , Male , Mice , Mice, Inbred C57BL
7.
Life Sci ; 286: 120067, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34678261

ABSTRACT

AIMS: Brain derived neurotrophic factor (BDNF) and the related receptors TrkB and p75NTR are expressed in skeletal muscle, yet their functions remain to be fully understood. Skeletal muscle denervation, which occurs in spinal injury, peripheral neuropathies, and aging, negatively affects muscle mass and function. In this study, we wanted to understand the role of BDNF, TrkB, and p75NTR in denervation-induced adverse effects on skeletal muscle. MAIN METHODS: Mice with unilateral sciatic denervation were used. Protein levels of pro- and mature BDNF, TrkB, p75NTR, activations of their downstream signaling pathways, and inflammation in the control and denervated muscle were measured with Western blot and tissue staining. Treatment with a p75NTR inhibitor and BDNF skeletal muscle specific knockout in mice were used to examine the role of p75NTR and pro-BDNF. KEY FINDINGS: In denervated muscle, pro-BDNF and p75NTR were significantly upregulated, and JNK and NF-kB, two major downstream signaling pathways of p75NTR, were activated, along with muscle atrophy and inflammation. Inhibition of p75NTR using LM11A-31 significantly reduced JNK activation and inflammatory cytokines in the denervated muscle. Moreover, skeletal muscle specific knockout of BDNF reduced pro-BDNF level, JNK activation and inflammation in the denervated muscle. SIGNIFICANCE: These results reveal for the first time that the upregulation of pro-BDNF and activation of p75NTR pathway are involved in denervation-induced inflammation in skeletal muscle. The results suggest that inhibition of pro-BDNF-p75NTR pathway can be a new target to treat skeletal muscle inflammation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Muscle, Skeletal/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Brain-Derived Neurotrophic Factor/physiology , Female , Isoleucine/analogs & derivatives , Isoleucine/pharmacology , Male , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Muscle Denervation/methods , Muscle, Skeletal/physiology , Muscular Atrophy/metabolism , Peripheral Nervous System Diseases , Protein Precursors/metabolism , Protein Precursors/physiology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Receptors, Nerve Growth Factor/physiology , Signal Transduction/physiology
8.
Neurobiol Aging ; 108: 146-154, 2021 12.
Article in English | MEDLINE | ID: mdl-34601245

ABSTRACT

Brain-derived neurotrophic factor (BNDF) plays a role in synapse integrity. We investigated in 398 cognitively normal adults (60±8years, 41% female, MMSE=28±1) the joint association of the Val66Met polymorphism of the BDNF gene (Met+/-) and plasma BDNF levels and abnormal cerebrospinal fluid (CSF) amyloid-beta status (A+/-) with cognitive decline and dementia risk. Age-, sex- and education-adjusted linear mixed models showed that compared to Met-A-, Met+A+ showed steeper decline on tests of global cognition, memory, language, attention and executive functioning, while Met-A+ showed steeper decline on a smaller number of tests. There were no associations between Met+A- and cognitive decline. Cox models showed that compared to Met-A-, Met+A+ participants were at increased risk of dementia (HR=8.8, 95%CI: 2.8-27.9), as were Met-A+ participants (HR=6.5, 95%CI: 2.2-19.5). Lower plasma BDNF was associated with an increased risk of progression to dementia in the A+ participants. Our results imply that Met-carriage on top of amyloid-beta pathology might increase rate of cognitive decline to dementia.


Subject(s)
Aging/genetics , Aging/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Brain-Derived Neurotrophic Factor/genetics , Cognition , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Polymorphism, Genetic , Aged , Aging/psychology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain-Derived Neurotrophic Factor/blood , Brain-Derived Neurotrophic Factor/physiology , Dementia/genetics , Dementia/metabolism , Female , Humans , Male , Middle Aged , Risk
9.
Mol Neurobiol ; 58(11): 5890-5906, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34415486

ABSTRACT

Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic ß-amyloid peptide (Aß). Aß induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aß to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aß deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aß, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Exercise , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Autophagy , Blood-Brain Barrier , Brain-Derived Neurotrophic Factor/physiology , Carrier Proteins/metabolism , Disease Models, Animal , Exercise/physiology , Fibronectins/physiology , Glymphatic System , Humans , Membrane Microdomains/physiology , Mice , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/prevention & control , Neuroinflammatory Diseases/physiopathology , Peptide Hydrolases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Physical Conditioning, Animal , Proteolysis , Signal Transduction/physiology , Sirtuin 1/physiology , Unfolded Protein Response/physiology
10.
Mol Biol Rep ; 48(6): 5249-5257, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34251558

ABSTRACT

Brain-derived neurotrophic factor (BDNF) enhances periodontal tissue regeneration. Tissue regeneration is characterized by inflammation, which directs the quality of tissue repair. This study aimed to investigate the effect of BDNF on the phagocytic activity of RAW264.7 cells. In addition, we studied the effect of BDNF on guanosine triphosphatase (GTP)-RAS-related C3 botulinus toxin substrate (Rac)1 and phospho-Rac1 levels in RAW264.7 cells. Rac1 inhibitor inhibited BDNF-induced phagocytosis of latex-beads. In addition, BDNF enhanced Porphyromonas gingivalis (Pg) phagocytosis by RAW264.7 cells as well as latex-beads. We demonstrated for the first time that BDNF enhances phagocytic activity of RAW264.7 cells through Rac1 activation. The present study proposes that BDNF may reduce inflammatory stimuli during BDNF-induced periodontal tissue regeneration through enhanced phagocytic activity of macrophages.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Macrophage Activation/genetics , Neuropeptides/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Brain-Derived Neurotrophic Factor/physiology , Cell Line , Guided Tissue Regeneration, Periodontal/methods , Inflammation , Macrophages/metabolism , Mice , Neuropeptides/physiology , Phagocytosis/physiology , Porphyromonas gingivalis/pathogenicity , RAW 264.7 Cells , rac1 GTP-Binding Protein/physiology
11.
J Integr Neurosci ; 20(2): 287-296, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34258927

ABSTRACT

A video-oculographic interface is a system for controlling objects using eye movements. The video-oculographic interface differs from other brain-computer interfaces regarding its improved accuracy, simplicity, and ergonomics. Despite these advantages, all users are not equally successful in mastering these various devices. It has been suggested that the genetic characteristics of the operators may determine the efficiency of video-oculographic interface mastery. We recruited healthy users with rs6313, rs2030324, rs429358, rs10119, rs457062, rs4290270, and rs6265 polymorphisms and analyzed the relationships between these polymorphisms and values of success in video-oculographic interface mastery. We found that carriers of the G/G genotype of the rs6265 polymorphism (BDNF gene) demonstrated the best results in video-oculographic interface mastery. In contrast, carriers of the A/A genotype were characterized by large standard deviations in the average amplitude of eye movement and the range of eye movement negatively correlated with goal achievement. This can be explained through the fact that carriers of the A/A genotype demonstrate lower synaptic plasticity due to reduced expression of BDNF when compared to carriers of the G/G genotype. These results expand our understanding of the genetic predictors of successful video-oculographic interface management, which will help to optimize device management training for equipment operators and people with disabilities.


Subject(s)
Brain-Computer Interfaces , Brain-Derived Neurotrophic Factor/physiology , Eye-Tracking Technology , Psychomotor Performance/physiology , Adult , Brain-Derived Neurotrophic Factor/genetics , Female , Humans , Male , Polymorphism, Single Nucleotide , Young Adult
12.
PLoS Biol ; 19(7): e3001337, 2021 07.
Article in English | MEDLINE | ID: mdl-34292944

ABSTRACT

Peripheral nerve injury-induced mechanical allodynia is often accompanied by abnormalities in the higher cortical regions, yet the mechanisms underlying such maladaptive cortical plasticity remain unclear. Here, we show that in male mice, structural and functional changes in the primary somatosensory cortex (S1) caused by peripheral nerve injury require neuron-microglial signaling within the local circuit. Following peripheral nerve injury, microglia in the S1 maintain ramified morphology and normal density but up-regulate the mRNA expression of brain-derived neurotrophic factor (BDNF). Using in vivo two-photon imaging and Cx3cr1CreER;Bdnfflox mice, we show that conditional knockout of BDNF from microglia prevents nerve injury-induced synaptic remodeling and pyramidal neuron hyperactivity in the S1, as well as pain hypersensitivity in mice. Importantly, S1-targeted removal of microglial BDNF largely recapitulates the beneficial effects of systemic BDNF depletion on cortical plasticity and allodynia. Together, these findings reveal a pivotal role of cerebral microglial BDNF in somatosensory cortical plasticity and pain hypersensitivity.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Brain/metabolism , Hyperalgesia/physiopathology , Microglia/metabolism , Neuronal Plasticity/physiology , Peripheral Nerve Injuries/metabolism , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Mice , Mice, Knockout , Peripheral Nerve Injuries/physiopathology
13.
Am J Physiol Endocrinol Metab ; 321(1): E164-E168, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34056921

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease that is characterized by progressive declines in cognitive function. Current epidemiological data indicate significant sex-linked disparities, where females have a higher risk of developing AD compared with male counterparts. This disparity necessitates further investigations to uncover the pathological and molecular factors influencing these sex differences. Although the underlying pathways behind this observed disparity remain elusive, recent research points to menopausal estrogen loss as a potential factor. Estrogen holds a significant role in amyloid precursor protein (APP) processing and overall neuronal health through the regulation of brain-derived neurotrophic factor (BDNF), a factor that is also reduced in postmenopausal women. BDNF is a known contributor to neuronal health and its reduced expression is typically linked to AD disorders. Exercise is known to increase BDNF and may provide an accessible activity for postmenopausal women to reduce their risk of AD. This review aims to discuss the relationship between estrogen, exercise, and BDNF in AD pathology.


Subject(s)
Alzheimer Disease , Estrogens/physiology , Exercise/physiology , Alzheimer Disease/epidemiology , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/metabolism , Brain-Derived Neurotrophic Factor/physiology , Female , Humans , Male , Menopause , Neurons/physiology , Risk Factors , Sex Factors
14.
Theranostics ; 11(13): 6154-6172, 2021.
Article in English | MEDLINE | ID: mdl-33995651

ABSTRACT

SH2 domain containing tyrosine phosphatase 2 (Shp2; PTPN11) regulates several intracellular pathways downstream of multiple growth factor receptors. Our studies implicate that Shp2 interacts with Caveolin-1 (Cav-1) protein in retinal ganglion cells (RGCs) and negatively regulates BDNF/TrkB signaling. This study aimed to investigate the mechanisms underlying the protective effects of shp2 silencing in the RGCs in glaucomatous conditions. Methods: Shp2 was silenced in the Cav-1 deficient mice and the age matched wildtype littermates using adeno-associated viral (AAV) constructs. Shp2 expression modulation was performed in an acute and a chronic mouse model of experimental glaucoma. AAV2 expressing Shp2 eGFP-shRNA under a strong synthetic CAG promoter was administered intravitreally in the animals' eyes. The contralateral eye received AAV-eGFP-scramble-shRNA as control. Animals with Shp2 downregulation were subjected to either microbead injections or acute ocular hypertension experimental paradigm. Changes in inner retinal function were evaluated by measuring positive scotopic threshold response (pSTR) while structural and biochemical alterations were evaluated through H&E staining, western blotting and immunohistochemical analysis of the retinal tissues. Results: A greater loss of pSTR amplitudes was observed in the WT mice compared to Cav-1-/- retinas in both the models. Silencing of Shp2 phosphatase imparted protection against inner retinal function loss in chronic glaucoma model in WT mice. The functional rescue also translated to structural preservation of ganglion cell layer in the chronic glaucoma condition in WT mice which was not evident in Cav-1-/- mice retinas. Conclusions: This study indicates that protective effects of Shp2 ablation under chronic experimental glaucoma conditions are dependent on Cav-1 in the retina, suggesting in vivo interactions between the two proteins.


Subject(s)
Caveolin 1/physiology , Genetic Therapy , Genetic Vectors/therapeutic use , Glaucoma/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Retina/pathology , Alpha-Globulins/genetics , Animals , Apoptosis , Brain-Derived Neurotrophic Factor/physiology , Caveolin 1/deficiency , Caveolin 1/genetics , DNA, Complementary/genetics , Dependovirus/genetics , Focal Adhesion Kinase 1/physiology , Gene Knockdown Techniques , Genes, Reporter , Genes, Synthetic , Glaucoma/metabolism , Glaucoma/pathology , Integrin beta1/physiology , Intraocular Pressure , Intravitreal Injections , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein-Tyrosine Kinases/physiology , Up-Regulation
15.
Brain Res ; 1767: 147536, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34052261

ABSTRACT

Intracerebral hemorrhage (ICH) is a subtype of stroke that causes major motor impairments. Brain-derived neurotrophic factor (BDNF) is known to have important roles in neuroplasticity and beneficially contributes to stroke recovery. This study aimed to characterize BDNF expression in the motor cortex after ICH and investigate the relationship between cortical BDNF expression and behavioral outcomes using an ICH rat model. Wistar rats were divided into two groups: a SHAM group (n = 7) and an ICH group (n = 8). ICH was induced by the injection of collagenase into the left striatum near the internal capsule. For behavioral assessments, the cylinder test and open field test were performed before surgery and 3 days, 1 week, 2 weeks, and 4 weeks after surgery. Following the behavioral assessments at 4 weeks, BDNF expression in the ipsilateral and contralateral motor cortex was assayed using RT-PCR and ELISA methods. There was no significant difference in either cortical BDNF mRNA or protein expression levels between the SHAM and ICH groups. However, the asymmetry index of BDNF mRNA expression between the ipsilateral and contralateral hemispheres shifted to the ipsilateral hemisphere after ICH. Furthermore, the ipsilateral cortical BDNF mRNA expression level positively correlated with motor function in the affected forelimb after ICH. This study describes for the first time that cortical BDNF mRNA expression is related to post-ICH motor impairment. These results highlight the importance of assessing the interhemispheric laterality of BDNF expression and could help develop novel treatment strategies for BDNF-dependent recovery after ICH.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Motor Activity/genetics , Motor Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/physiology , Cerebral Hemorrhage/metabolism , Disease Models, Animal , Exercise Therapy/methods , Forelimb/metabolism , Functional Laterality/physiology , Gene Expression/genetics , Gene Expression Regulation/genetics , Male , Motor Activity/physiology , Motor Cortex/physiology , Neuronal Plasticity , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recovery of Function , Transcriptome/genetics
16.
Int Urol Nephrol ; 53(10): 2177-2187, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33881703

ABSTRACT

PURPOSE: Diabetic nephropathy (DN) is one of the most serious complications of diabetes that leads to decline of renal function. Although numerous studies have revealed that microRNAs (miRNAs) play essential roles in the progression of DN, whether miR-365 is involved remains elusive. METHODS: The successful construction of DN model was confirmed by ELSIA, hematoxylin-eosin (HE) and Masson staining assay. The expression of miR-365 was detected through RT-qPCR. The levels of BDNF, p-TrkB, α-smooth muscle actin (SMA), collagen IV (Col.IV), transforming growth factor-ß1 (TGF-ß1), tumor necrosis factor α (TNF-α), and interleukin-6 (IL-6) were evaluated by western blot, IF or ELISA assays. Luciferase reporter assay was used to detect the interaction between miR-365 and BDNF. RESULTS: The DN mice model was induced by streptozotocin (STZ). Then miR-365 expression was found to upregulate in tissues of DN rat. Furthermore, elevated expression of miR-365 was found in high glucose (HG)-treated HK-2 cells. Silencing of miR-365 suppressed the accumulation of ECM components and secretion of inflammatory cytokines in HK-2 cells. In addition, it was demonstrated that miR-365 could target BDNF. The protein levels of BDNF and p-TrkB were negatively regulated by miR-365 in HK-2 cells. Moreover, inhibition of miR-365 suppressed the levels of SMA, Col.IV, TGF-ß1, TNF-α, and IL-6, indicating the renal fibrosis was inhibited by miR-365 knockdown. CONCLUSION: MiR-365 could regulate BDNF-TrkB signal axis in STZ induced DN fibrosis and renal function. The results of the current study might provide a promising biomarker for the treatment of DN in the future.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/complications , Diabetic Nephropathies/physiopathology , Kidney/pathology , Kidney/physiopathology , Membrane Glycoproteins/physiology , MicroRNAs/physiology , Protein-Tyrosine Kinases/physiology , Animals , Cells, Cultured , Fibrosis/etiology , Humans , Male , Mice , Rats , Rats, Sprague-Dawley
17.
Neuropeptides ; 87: 102135, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33812160

ABSTRACT

BACKGROUND & AIMS: Schizophrenia is a serious long-term disorder in which the metabolic complications and abnormalities of the brain-derived neurotrophic factor (BDNF) can be found. In this study, we conducted a systematic review of the relationship between BDNF, metabolic syndrome (MetS) and its components in schizophrenic patients. METHODS: Data were collected mainly from PubMed, Google Scholar, Scopus, and ProQuest databases. The keywords related to the BDNF, MetS, schizophrenia were searched. Two reviewers independently screened 1061 abstracts. And eventually, a total of 7 studies (6 observational and 1 interventional) was included in the systematic reviews. RESULTS: Four of the 7 study ascertained statistically significant inverse relationship between serum BDNF levels and MetS in schizophrenic patients. While in the other two studies, there was no inverse relationship. In the last selected study, the researchers found a weak association between the Val66Met polymorphism in BDNF Gene and clozapine-induced MetS. CONCLUSION: Although this relationship could not be determined but BDNF levels appear to be reduced in schizophrenic patients with MetS and factors such as sex and antipsychotic class differentiation, sampling and methodology and episodes of illness could play a role in the results and outcomes.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Metabolic Syndrome/blood , Schizophrenia/blood , Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Chronic Disease , Clinical Trials as Topic , Clozapine/adverse effects , Clozapine/therapeutic use , Comorbidity , Diet , Fasting/adverse effects , Female , Genetic Predisposition to Disease , Humans , Hypertriglyceridemia/chemically induced , Life Style , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/epidemiology , Metabolic Syndrome/genetics , Mutation, Missense , Observational Studies as Topic , Polymorphism, Single Nucleotide , Prevalence , Schizophrenia/drug therapy , Schizophrenia/epidemiology , Sex Factors
18.
Mol Nutr Food Res ; 65(9): e2000869, 2021 05.
Article in English | MEDLINE | ID: mdl-33783973

ABSTRACT

INTRODUCTION: Degraded porphyran is a bioactive polysaccharide extracted from Porphyra haitanensis (P. haitanensis). According to the previous studies, it produced anti-inflammatory activity, but little is known about its effects on depression. METHODS AND RESULTS: As inflammation is one of the critical factors involved in the development of depression, this study aims to elucidate the potential antidepressant-like effects of degraded porphyran. The results show that acute porphyran treatment decreased the immobility time in despair tests. In addition, subchronic porphyran administration reverses depressive-like behaviors in lipopolysaccharide (LPS)-treated mice. Meanwhile, porphyran inhibits NF-κB/NLRP3 signaling, proinflammatory cytokine release, and microglial activation in the hippocampus. Moreover, chronic porphyran treatment activates hippocampal brain derived neurotrophic factor (BDNF)/TrkB/ERK/CREB signaling pathway in chronic unpredictable mild stress (CUMS) in mice. As a result, neurogenesis and spinogenesis are maintained. CONCLUSIONS: The findings of the present study indicate that degraded porphyran intake provides a potential strategy for depression treatment, which is mediated by the inhibition of neuroinflammation and the enhancement of neurogenesis and spinogenesis in the central nervous systems.


Subject(s)
Antidepressive Agents/pharmacology , Porphyra/chemistry , Sepharose/analogs & derivatives , Animals , Brain-Derived Neurotrophic Factor/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Molecular Docking Simulation , Neurogenesis/drug effects , Neuroinflammatory Diseases/drug therapy , Sepharose/pharmacology , Toll-Like Receptor 4/physiology
19.
Pediatr Neurol ; 118: 35-39, 2021 05.
Article in English | MEDLINE | ID: mdl-33773288

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, has an extensively studied classical role in neuronal growth, differentiation, survival, and plasticity. Neurotrophic, from the Greek neuro and trophos, roughly translates as "vital nutrition for the brain." During development, BDNF and its associated receptor tyrosine receptor kinase B are tightly regulated as they influence the formation and maturation of neuronal synapses. Preclinical research investigating the role of BDNF in neurological disorders has focused on the effects of decreased BDNF expression on the development and maintenance of neuronal synapses. In contrast, heightened BDNF-tyrosine receptor kinase B activity has received less scrutiny for its role in neurological disorders. Recent studies suggest that excessive BDNF-tyrosine receptor kinase B signaling in the developing brain may promote the hyperexcitability that underlies refractory neonatal seizures. This review will critically examine BDNF-tyrosine receptor kinase B signaling in the immature brain, its role in the emergence of refractory neonatal seizures, and the potential of targeting BDNF-TrkB signaling as a novel antiseizure strategy.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Seizures/etiology , Seizures/therapy , Humans , Infant, Newborn , Membrane Glycoproteins/physiology , Receptor, trkB/physiology , Seizures/metabolism , Signal Transduction/physiology
20.
J Neurochem ; 158(2): 169-181, 2021 07.
Article in English | MEDLINE | ID: mdl-33742683

ABSTRACT

Morphine withdrawal evokes neuronal apoptosis through mechanisms that are still under investigation. We have previously shown that morphine withdrawal increases the levels of pro-brain-derived neurotrophic factor (BDNF), a proneurotrophin that promotes neuronal apoptosis through the binding and activation of the pan-neurotrophin receptor p75 (p75NTR). In this work, we sought to examine whether morphine withdrawal increases p75NTR-driven signaling events. We employed a repeated morphine treatment-withdrawal paradigm in order to investigate biochemical and histological indicators of p75NTR-mediated neuronal apoptosis in mice. We found that repeated cycles of spontaneous morphine withdrawal promote an accumulation of p75NTR in hippocampal synapses. At the same time, TrkB, the receptor that is crucial for BDNF-mediated synaptic plasticity in the hippocampus, was decreased, suggesting that withdrawal alters the neurotrophin receptor environment to favor synaptic remodeling and apoptosis. Indeed, we observed evidence of neuronal apoptosis in the hippocampus, including activation of c-Jun N-terminal kinase (JNK) and increased active caspase-3. These effects were not seen in saline or morphine-treated mice which had not undergone withdrawal. To determine whether p75NTR was necessary in promoting these outcomes, we repeated these experiments in p75NTR heterozygous mice. The lack of one p75NTR allele was sufficient to prevent the increases in phosphorylated JNK and active caspase-3. Our results suggest that p75NTR participates in the neurotoxic and proinflammatory state evoked by morphine withdrawal. Because p75NTR activation negatively influences synaptic repair and promotes cell death, preventing opioid withdrawal is crucial for reducing neurotoxic mechanisms accompanying opioid use disorders.


Subject(s)
Apoptosis , Morphine Dependence/pathology , Neurons/pathology , Receptors, Nerve Growth Factor/metabolism , Substance Withdrawal Syndrome/pathology , Alleles , Animals , Apoptosis/physiology , Brain-Derived Neurotrophic Factor/physiology , Caspase 3/metabolism , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...