Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.457
Filter
1.
Sci Signal ; 17(855): eadn2616, 2024 09 24.
Article in English | MEDLINE | ID: mdl-39316663

ABSTRACT

Basal forebrain cholinergic neurons (BFCNs) extend long projections to multiple regions in the brain to regulate cognitive functions. Degeneration of BFCNs is seen with aging, after brain injury, and in neurodegenerative disorders. An increase in the amount of the immature proform of nerve growth factor (proNGF) in the cerebral cortex results in retrograde degeneration of BFCNs through activation of proNGF receptor p75NTR. Here, we investigated the signaling cascades initiated at the axon terminal that mediate proNGF-induced retrograde degeneration. We found that local axonal protein synthesis and retrograde transport mediated proNGF-induced degeneration initiated from the axon terminal. Analysis of the nascent axonal proteome revealed that proNGF stimulation of axonal terminals triggered the synthesis of numerous proteins within the axon, and pathway analysis showed that amyloid precursor protein (APP) was a key upstream regulator in cultured BFCNs and in mice. Our findings reveal a functional role for APP in mediating BFCN axonal degeneration and cell death induced by proNGF.


Subject(s)
Amyloid beta-Protein Precursor , Axons , Basal Forebrain , Nerve Growth Factor , Animals , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Axons/metabolism , Axons/pathology , Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Basal Forebrain/metabolism , Basal Forebrain/pathology , Mice , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Protein Precursors/metabolism , Protein Precursors/genetics , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Mice, Inbred C57BL , Cells, Cultured , Signal Transduction
2.
Int J Biol Macromol ; 279(Pt 4): 135376, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39244119

ABSTRACT

Adipocyte-secreted factors intricately regulate adipose tissue function, and the underlying molecular mechanisms are only partially understood. However, the function of PRELP, which is a key component of the extracellular matrix (ECM) in adipocytes, remains largely unknown. In this study, we demonstrate that PRELP was upregulated in both obese humans and mice, which exhibited a positive correlation with metabolic disorders. PRELP knockout could resist HFD-induced obesity and inhibit adipocyte differentiation. PRELP knockout improved glucose tolerance, insulin sensitivity and alleviated adipose tissue fibrosis. Mechanistically, PRELP was secreted into the ECM and bound to the extracellular domain of its receptor p75NTR in adipocytes, which further activated the FAK/MAPK (JNK, p38 MAPK, ERK1/2) signaling pathway, promoting adipocyte differentiation and exacerbating adipocyte fibrosis. Adipocyte PRELP plays a pivotal role in regulating obesity and adipose tissue fibrosis through an autocrine manner, and PRELP may be a therapeutic target for obesity and its related metabolic disorders.


Subject(s)
Adipocytes , Adipose Tissue , Fibrosis , MAP Kinase Signaling System , Animals , Mice , Humans , Adipose Tissue/metabolism , Adipocytes/metabolism , Cell Differentiation , Obesity/metabolism , Obesity/pathology , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Male , Mice, Knockout , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Protein Binding , Adipogenesis , Mice, Inbred C57BL , Nerve Tissue Proteins
3.
Cell Genom ; 4(9): 100642, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39216475

ABSTRACT

Genetic variants in ABCA7, an Alzheimer's disease (AD)-associated gene, elevate AD risk, yet its functional relevance to the etiology is unclear. We generated a CRISPR-Cas9-mediated abca7 knockout zebrafish to explore ABCA7's role in AD. Single-cell transcriptomics in heterozygous abca7+/- knockout combined with Aß42 toxicity revealed that ABCA7 is crucial for neuropeptide Y (NPY), brain-derived neurotrophic factor (BDNF), and nerve growth factor receptor (NGFR) expressions, which are crucial for synaptic integrity, astroglial proliferation, and microglial prevalence. Impaired NPY induction decreased BDNF and synaptic density, which are rescuable with ectopic NPY. In induced pluripotent stem cell-derived human neurons exposed to Aß42, ABCA7-/- suppresses NPY. Clinical data showed reduced NPY in AD correlated with elevated Braak stages, genetic variants in NPY associated with AD, and epigenetic changes in NPY, NGFR, and BDNF promoters linked to ABCA7 variants. Therefore, ABCA7-dependent NPY signaling via BDNF-NGFR maintains synaptic integrity, implicating its impairment in increased AD risk through reduced brain resilience.


Subject(s)
Alzheimer Disease , Brain-Derived Neurotrophic Factor , Neuropeptide Y , Signal Transduction , Zebrafish , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Neuropeptide Y/metabolism , Neuropeptide Y/genetics , Humans , Synapses/metabolism , Synapses/pathology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics
4.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125882

ABSTRACT

Neurotrophins and their receptors are distinctly expressed during brain development and play crucial roles in the formation, survival, and function of neurons in the nervous system. Among these molecules, brain-derived neurotrophic factor (BDNF) has garnered significant attention due to its involvement in regulating GABAergic system development and function. In this review, we summarize and compare the expression patterns and roles of neurotrophins and their receptors in both the developing and adult brains of rodents, macaques, and humans. Then, we focus on the implications of BDNF in the development and function of GABAergic neurons from the cortex and the striatum, as both the presence of BDNF single nucleotide polymorphisms and disruptions in BDNF levels alter the excitatory/inhibitory balance in the brain. This imbalance has different implications in the pathogenesis of neurodevelopmental diseases like autism spectrum disorder (ASD), Rett syndrome (RTT), and schizophrenia (SCZ). Altogether, evidence shows that neurotrophins, especially BDNF, are essential for the development, maintenance, and function of the brain, and disruptions in their expression or signaling are common mechanisms in the pathophysiology of brain diseases.


Subject(s)
Brain-Derived Neurotrophic Factor , GABAergic Neurons , Humans , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , GABAergic Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Brain/metabolism , Brain/growth & development
5.
J Neurol Sci ; 464: 123169, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39126731

ABSTRACT

BACKGROUND: Brain recovery mechanisms after injuries like aneurysmal subarachnoid hemorrhage (aSAH), ischemic stroke (IS), and traumatic brain injury (TBI) involve brain plasticity, synaptic regeneration, and neuroinflammation. We hypothesized that serum levels of the p75 neurotrophic receptor (p75NTR) and associated signaling proteins, as well as differentially expressed (DE) microRNAs, could predict recovery outcomes irrespective of injury type. METHODS: A prospective patient cohort with ischemic stroke (IS, n = 30), aneurysmal subarachnoid hemorrhage (aSAH, n = 31), and traumatic brain injury (TBI, n = 13) were evaluated (total n = 74). Serum samples were collected at two post-injury intervals (early: 1-3 days, late: 4-8 days), and outcomes were assessed after three months using the modified Rankin Scale (mRS), categorizing outcomes as favorable (mRS 0-3) or unfavorable (mRS 4-6). Six proteins were measured using ELISAs: p75NTR, NGF, sortilin, IL1ß, TNFα, and cyclophilin. DE microRNAs were identified using DESeq2, and their target genes were predicted. Serum molecules between patients with differing outcomes were compared using a Kolmogorov-Smirnov test, 2-tailed t-test and multivariate linear discriminant analysis (LDA). RESULTS: Favorable (n = 46) and unfavorable (n = 28) outcome cohorts were balanced with age and sex (p = 0.25 and 0.63). None of the studied proteins correlated with age. Combinatory LDA of the six protein biomarkers indicated strong prognostic value for favorable outcomes (OR 2.09; AUC = 70.3%, p = 0.0058). MicroRNA expression changes over time were identified in the aSAH, TBI, and IS groups (p < 0.05, FDR corrected). Twenty-three microRNAs were commonly DE across all brain injury groups when comparing favorable and unfavorable outcomes (p < 0.05). LDA of four microRNAs targeting the studied proteins showed high prognostic accuracy (OR 11.7; AUC = 94.1%, p = 0.016). CONCLUSIONS: The combined prognostic microRNA and protein biomarker models demonstrated accurate outcome prognostication across diverse injury types, implying the presence of a common recovery mechanism. DE microRNAs were found to target the studied molecules, suggesting a potential mechanistic role in recovery. Further investigation is warranted to study these molecules in prognostication, as well as therapeutic targets for enhancing recovery.


Subject(s)
Biomarkers , Circulating MicroRNA , Neuronal Plasticity , Humans , Male , Female , Middle Aged , Prospective Studies , Biomarkers/blood , Circulating MicroRNA/blood , Aged , Neuronal Plasticity/physiology , Adult , Subarachnoid Hemorrhage/blood , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/diagnosis , Cohort Studies , Neuroinflammatory Diseases/blood , Ischemic Stroke/blood , Receptors, Nerve Growth Factor/blood , Receptors, Nerve Growth Factor/genetics , Recovery of Function/physiology , Prognosis , Nerve Tissue Proteins , Adaptor Proteins, Vesicular Transport
6.
Cells ; 13(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39120279

ABSTRACT

RESEARCH QUESTION: Theca interna cells (TICs) are an indispensable cell source for ovarian follicle development and steroidogenesis. Recent studies have identified theca stem cells (TSCs) in both humans and animals. Interestingly, TSCs express mesenchymal stem cell (MSC)-related markers and can differentiate into mesenchymal lineages. MSCs are promising for tissue engineering and regenerative medicine due to their self-renewal and differentiation abilities. Therefore, this study investigated the potential origin of TICs from MSCs. DESIGN: Whole ovaries from postmenopausal organ donors were obtained, and their cortex was cryopreserved prior to the isolation of stromal cells. These isolated cells were differentiated in vitro to TICs using cell media enriched with various growth factors and hormones. Immunocytochemistry, an enzyme-linked immunosorbent assay, flow cytometry, and reverse transcription-quantitative polymerase chain were employed at different timepoints. Data were analyzed using one-way ANOVA. RESULTS: Immunocytochemistry showed an increase in TIC markers from day 0 to day 8 and a significant rise in MSC-like markers on day 2. This corresponds with rising androstenedione levels from day 2 to day 13. Flow cytometry identified a decreasing MSC-like cell population from day 2 onwards. The CD13+ cell population and its gene expression increased significantly over time. NGFR and PDGFRA expression was induced on days 0 and 2, respectively, compared to day 13. CONCLUSIONS: This study offers insights into MSC-like cells as the potential origin of TICs. Differentiating TICs from these widely accessible MSCs holds potential significance for toxicity studies and investigating TIC-related disorders like polycystic ovary syndrome (PCOS).


Subject(s)
Cell Differentiation , Theca Cells , Female , Theca Cells/metabolism , Theca Cells/cytology , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Cells, Cultured , Biomarkers/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics
7.
CNS Neurosci Ther ; 30(7): e14875, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39072998

ABSTRACT

BACKGROUND: Angiogenesis is crucial in neuroprotection of secondary thalamic injury after cortical infarction. The p75 neurotrophin receptor (p75NTR) plays a key role in activating angiogenesis. However, the effects of p75NTR on angiogenesis in the thalamus after cortical infarction are largely unknown. Herein we investigate whether p75NTR facilitates angiogenesis to attenuate secondary thalamic damage via activating hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway mediated by Von Hippel-Lindau (VHL) after distal middle cerebral artery occlusion (dMCAO). METHODS: The male rat model of dMCAO was established. The effects of p75NTR on the angiogenesis was evaluated using RNA-sequencing, immunohistochemistry, western blot, quantitative real-time polymerase chain reaction, magnetic resonance imaging, behavior tests, viral and pharmacological interventions. RESULTS: We found that the p75NTR and vessel density were decreased in ipsilateral thalamus after dMCAO. The p75NTR-VHL interaction was reduced, which promoted the ubiquitination degradation of HIF-1α and reduced VEGF expression after dMCAO. Notably, p75NTR overexpression restrained the ubiquitination degradation of HIF-1α by inhibiting VHL-HIF-1α interaction, further promoted angiogenesis, increased cerebral blood flow of ipsilateral thalamus and improved neurological function after dMCAO. CONCLUSION: For the first time, we highlighted that the enhancement of p75NTR-VHL interaction promoted angiogenesis in attenuating secondary thalamic damage after dMCAO.


Subject(s)
Infarction, Middle Cerebral Artery , Neovascularization, Physiologic , Rats, Sprague-Dawley , Thalamus , Animals , Male , Rats , Thalamus/metabolism , Thalamus/pathology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Neovascularization, Physiologic/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Cerebral Infarction/pathology , Angiogenesis , Nerve Tissue Proteins , Receptors, Growth Factor
8.
Transgenic Res ; 33(4): 255-266, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38981975

ABSTRACT

The p75NTR neurotrophin receptor has positive and negative roles regulating cell survival in the nervous system. Unambiguous interpretation of p75NTR function in vivo has been complicated, however, by residual expression of alternate forms of p75NTR protein in initial p75NTR knock-out mouse models. As rats are the preferred rodent for studying brain and behaviour, and to simplify interpretation of the knock-out phenotype, we report here the generation of a mutant rat devoid of the p75NTR protein. TALEN-mediated recombination in embryonic stem cells (ESCs) was used to flank exon 2 of p75NTR with Lox P sites and produce transgenic rats carrying either un-recombined floxed p75NTREx2-fl, or recombined, exon-2 deleted p75NTREx2-Δ alleles. Crossing p75NTREx2-fl rats with a Cre-deleter strain efficiently removed exon 2 in vivo. Excision of exon 2 causes a frameshift after p75NTR Gly23 and eliminated p75NTR protein expression. Rats lacking p75NTR were healthy, fertile, and histological analysis did not reveal significant changes in cellular density or overall structure in their brains. p75NTR function is therefore largely dispensable for normal development, growth and basal homeostasis in the rat. However, the availability of constitutive and conditional p75NTREx2-Δ rats provides new opportunities to investigate specific roles of p75NTR upon injury and during tissue repair.


Subject(s)
Rats, Transgenic , Animals , Rats , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Fertility/genetics , Female , Brain/metabolism , Brain/growth & development , Male , Exons/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Embryonic Stem Cells/metabolism , Receptors, Growth Factor
9.
Cancer Sci ; 115(7): 2346-2359, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710200

ABSTRACT

RNAs, such as noncoding RNA, microRNA, and recently mRNA, have been recognized as signal transduction molecules. CD271, also known as nerve growth factor receptor, has a critical role in cancer, although the precise mechanism is still unclear. Here, we show that CD271 mRNA, but not CD271 protein, facilitates spheroid cell proliferation. We established CD271-/- cells lacking both mRNA and protein of CD271, as well as CD271 protein knockout cells lacking only CD271 protein, from hypopharyngeal and oral squamous cell carcinoma lines. Sphere formation was reduced in CD271-/- cells but not in CD271 protein knockout cells. Mutated CD271 mRNA, which is not translated to a protein, promoted sphere formation. CD271 mRNA bound to hnRNPA2B1 protein at the 3'-UTR region, and the inhibition of this interaction reduced sphere formation. In surgical specimens, the CD271 mRNA/protein expression ratio was higher in the cancerous area than in the noncancerous area. These data suggest CD271 mRNA has dual functions, encompassing protein-coding and noncoding roles, with its noncoding RNA function being predominant in oral and head and neck squamous cell carcinoma.


Subject(s)
Head and Neck Neoplasms , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Mouth Neoplasms , Nerve Tissue Proteins , RNA, Messenger , Receptors, Nerve Growth Factor , Squamous Cell Carcinoma of Head and Neck , Female , Humans , Male , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism
10.
Clin Immunol ; 263: 110206, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599263

ABSTRACT

Patients suffering from sepsis-induced acute lung injury (ALI) exhibit a high mortality rate, and their prognosis is closely associated with infiltration of neutrophils into the lungs. In this study, we found a significant elevation of CD64+ neutrophils, which highly expressed p75 neurotrophin receptor (p75NTR) in peripheral blood of mice and patients with sepsis-induced ALI. p75NTR+CD64+ neutrophils were also abundantly expressed in the lung of ALI mice induced by lipopolysaccharide. Conditional knock-out of the myeloid lineage's p75NTR gene improved the survival rates, attenuated lung tissue inflammation, reduced neutrophil infiltration and enhanced the phagocytic functions of CD64+ neutrophils. In vitro, p75NTR+CD64+ neutrophils exhibited an upregulation and compromised phagocytic activity in blood samples of ALI patients. Blocking p75NTR activity by soluble p75NTR extracellular domain peptide (p75ECD-Fc) boosted CD64+ neutrophils phagocytic activity and reduced inflammatory cytokine production via regulation of the NF-κB activity. The findings strongly indicate that p75NTR+CD64+ neutrophils are a novel pathogenic neutrophil subpopulation promoting sepsis-induced ALI.


Subject(s)
Acute Lung Injury , Mice, Inbred C57BL , Neutrophils , Phagocytosis , Receptors, IgG , Receptors, Nerve Growth Factor , Sepsis , Animals , Acute Lung Injury/immunology , Acute Lung Injury/etiology , Neutrophils/immunology , Neutrophils/metabolism , Sepsis/immunology , Sepsis/complications , Humans , Receptors, IgG/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology , Mice , Male , Phagocytosis/immunology , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/immunology , Mice, Knockout , Lipopolysaccharides , Cytokines/metabolism , Cytokines/immunology , Lung/immunology , Lung/pathology , Female , NF-kappa B/metabolism , NF-kappa B/immunology , Nerve Tissue Proteins
11.
FASEB J ; 38(1): e23312, 2024 01.
Article in English | MEDLINE | ID: mdl-38161282

ABSTRACT

ProBDNF is the precursor protein of brain-derived neurotrophic factor (BDNF) expressed in the central nervous system and peripheral tissues. Previous studies showed that the blood levels of both proBDNF and p75 neurotrophic receptors (p75NTR) in major depressive disorder (MDD) were increased, but which blood cell types express proBDNF and its receptors is not known. Furthermore, the relationship between proBDNF/p75NTR and inflammatory cytokines in peripheral blood of MDD is unclear. Peripheral blood mononuclear cells (PBMCs) and serum were obtained from depressive patients (n = 32) and normal donors (n = 20). We examined the expression of proBDNF and inflammatory markers and their correlative relationship in patients with major depression. Using flow cytometry analysis, we examined which blood cells express proBDNF and its receptors. Finally, the role of proBDNF/p75NTR signal in inflammatory immune activity of PBMCs was verified in vitro experiments. Inflammatory cytokines in PBMC from MDD patients were increased and correlated with the major depression scores. The levels of IL-1ß and IL-10 were also positively correlated with the major depression scores, while the levels of TNF-α and IL-6 were negatively correlated with the major depression scores. Intriguingly, the levels of sortilin were positively correlated with IL-1ß. Q-PCR and Western blots showed proBDNF, p75NTR, and sortilin levels were significantly increased in PBMCs from MDD patients compared with that from the normal donors. Flow cytometry studies showed that proBDNF and p75NTR were present mainly in CD4+ and CD8+ T cells. The number of proBDNF and p75NTR positive CD4+ and CD8+ T cells from MDD patients was increased and subsequently reversed after therapeutic management. Exogenous proBDNF protein or p75ECD-Fc treatment of cultured PBMC affected the release of inflammatory cytokines in vitro. ProBDNF promoted the expression of inflammatory cytokines, while p75ECD-Fc inhibited the expression of inflammatory cytokines. Given there was an inflammatory response of lymphocytes to proBDNF, it is suggested that proBDNF/p75NTR signaling may upstream inflammatory cytokines in MDD. Our data suggest that proBDNF/p75NTR signaling may not only serve as biomarkers but also may be a potential therapeutic target for MDD.


Subject(s)
Depressive Disorder, Major , Humans , Depressive Disorder, Major/metabolism , Leukocytes, Mononuclear/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Up-Regulation , CD8-Positive T-Lymphocytes/metabolism , Depression , Brain-Derived Neurotrophic Factor/metabolism , Cytokines/metabolism
12.
Genes Cells ; 29(1): 73-85, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38016691

ABSTRACT

Bladder cancer is a urothelial cancer and effective therapeutic strategies for its advanced stages are limited. Here, we report that CD271, a neurotrophin receptor, promotes the proliferation and migration of bladder cancer cells. CD271 knockdown decreased proliferation in both adherent and spheroid cultures, and vice versa when CD271 was overexpressed in bladder cancer cell lines. CD271 depletion impaired tumorigenicity in vivo. Migration activity was reduced by CD271 knockdown and TAT-Pep5, a known CD271-Rho GDI-binding inhibitor. Apoptosis was induced by CD271 knockdown. Comprehensive gene expression analysis revealed alterations in E2F- and Myc-related pathways upon CD271 expression. In clinical cases, patients with high CD271 expression showed significantly shortened overall survival. In surgically resected specimens, pERK, a known player in proliferation signaling, colocalizes with CD271. These data indicate that CD271 is involved in bladder cancer malignancy by promoting cell proliferation and migration, resulting in poor prognosis.


Subject(s)
Receptors, Nerve Growth Factor , Urinary Bladder Neoplasms , Humans , Adapalene , Receptors, Nerve Growth Factor/genetics , Cell Proliferation , Signal Transduction , Urinary Bladder Neoplasms/genetics , Cell Movement , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
13.
Mol Neurobiol ; 61(1): 276-293, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37606717

ABSTRACT

The precursor nerve growth factor (ProNGF) and its receptor p75 neurotrophin receptor (p75NTR) are upregulated in several brain diseases, including ischemic stroke. The activation of p75NTR is associated with neuronal apoptosis and inflammation. Thus, we hypothesized that p75NTR modulation attenuates brain damage and improves functional outcomes after ischemic stroke. Two sets of experiments were performed. (1) Adult wild-type (WT) C57BL/6 J mice were subjected to intraluminal suture-middle cerebral artery occlusion (MCAO) to induce cerebral ischemia. Pharmacological inhibitor of p75NTR, LM11A-31 (50 mg/kg), or normal saline was administered intraperitoneally (IP) 1 h post-MCAO, and animals survived for 24 h. (2) Adult p75NTR heterozygous knockout (p75NTR+/-) and WT were subjected to photothrombotic (pMCAO) to induce ischemic stroke, and the animals survived for 72 h. The sensory-motor function of animals was measured using Catwalk XT. The brain samples were collected to assess infarction volume, edema, hemorrhagic transformation, neuroinflammation, and signaling pathway at 24 and 72 h after the stroke. The findings described that pharmacological inhibition and genetic knocking down of p75NTR reduce infarction size, edema, and hemorrhagic transformation following ischemic stroke. Additionally, p75NTR modulation significantly decreased several anti-apoptosis markers and improved sensory motor function compared to the WT mice following ischemic stroke. Our observations exhibit that the involvement of p75NTR in ischemic stroke and modulation of p75NTR could improve the outcome of ischemic stroke by increasing cell survival and enhancing motor performance. LM11A-31 has the potential to be a promising therapeutic agent for ischemic stroke. However, more evidence is needed to illuminate the efficacy of LM11A-31 in ischemic stroke.


Subject(s)
Brain Injuries , Ischemic Stroke , Mice , Animals , Receptor, Nerve Growth Factor/metabolism , Ischemic Stroke/complications , Ischemic Stroke/drug therapy , Mice, Inbred C57BL , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Brain/metabolism , Infarction , Edema
14.
Int J Mol Sci ; 24(13)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37446129

ABSTRACT

In vertebrates, neurotrophins and their receptors play a fundamental role in the central and peripheral nervous systems. Several studies reported that each neurotrophin/receptor signalling pathway can perform various functions during axon development, neuronal growth, and plasticity. Previous investigations in some fish species have identified neurotrophins and their receptors in the spinal cord under physiological conditions and after injuries, highlighting their potential role during regeneration. In our study, for the first time, we used an excellent animal model, the zebrafish (Danio rerio), to compare the mRNA localization patterns of neurotrophins and receptors in the spinal cord. We quantified the levels of mRNA using qPCR, and identified the transcription pattern of each neurotrophin/receptor pathway via in situ hybridization. Our data show that ngf/trka are the most transcribed members in the adult zebrafish spinal cord.


Subject(s)
Nerve Growth Factors , Zebrafish , Animals , Nerve Growth Factors/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Receptors, Nerve Growth Factor/genetics , Spinal Cord/metabolism , RNA, Messenger/metabolism , Receptor, trkA/genetics
15.
Cell Death Dis ; 14(7): 440, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37460457

ABSTRACT

Cytosine arabinoside (AraC) is one of the main therapeutic treatments for several types of cancer, including acute myeloid leukaemia. However, after a high-dose AraC chemotherapy regime, patients develop severe neurotoxicity and cell death in the central nervous system leading to cerebellar ataxia, dysarthria, nystagmus, somnolence and drowsiness. AraC induces apoptosis in dividing cells. However, the mechanism by which it leads to neurite degeneration and cell death in mature neurons remains unclear. We hypothesise that the upregulation of the death receptor p75NTR is responsible for AraC-mediated neurodegeneration and cell death in leukaemia patients undergoing AraC treatment. To determine the role of AraC-p75NTR signalling in the cell death of mature neurons, we used mature cerebellar granule neurons' primary cultures from p75NTR knockout and p75NTRCys259 mice. Evaluation of neurite degeneration, cell death and p75NTR signalling was done by immunohistochemistry and immunoblotting. To assess the interaction between AraC and p75NTR, we performed cellular thermal shift and AraTM assays as well as Homo-FRET anisotropy imaging. We show that AraC induces neurite degeneration and programmed cell death of mature cerebellar granule neurons in a p75NTR-dependent manner. Mechanistically, Proline 252 and Cysteine 256 residues facilitate AraC interaction with the transmembrane domain of p75NTR resulting in uncoupling of p75NTR from the NFκB survival pathway. This, in turn, exacerbates the activation of the cell death/JNK pathway by recruitment of TRAF6 to p75NTR. Our findings identify p75NTR as a novel molecular target to develop treatments for counteract AraC-mediated cell death of mature neurons.


Subject(s)
Neurons , Receptors, Nerve Growth Factor , Animals , Mice , Apoptosis/physiology , Cell Death , Cells, Cultured , Neurites/metabolism , Neurons/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
16.
Methods Mol Biol ; 2681: 361-371, 2023.
Article in English | MEDLINE | ID: mdl-37405658

ABSTRACT

Suspension cells derived from human embryonic kidney cells (HEK 293) are attractive cell lines for retroviral vector production in gene therapeutic development studies and applications. The low-affinity nerve growth factor receptor (NGFR) is a genetic marker frequently used as a reporter gene in transfer vectors to detect and enrich genetically modified cells. However, the HEK 293 cell line and its derivatives endogenously express the NGFR protein. To eradicate the high background NGFR expression in future retroviral vector packaging cells, we here employed the CRISPR/Cas9 system to generate human suspension 293-F NGFR knockout cells. The expression of a fluorescent protein coupled via a 2A peptide motif to the NGFR targeting Cas9 endonuclease enabled the simultaneous depletion of cells expressing Cas9 and remaining NGFR-positive cells. Thus, a pure population of NGFR-negative 293-F cells lacking persistent Cas9 expression was obtained in a simple and easily applicable procedure.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , CRISPR-Cas Systems/genetics , Gene Editing/methods , Receptor, Nerve Growth Factor/genetics , HEK293 Cells , Genetic Vectors/genetics , Receptors, Nerve Growth Factor/genetics , Nerve Tissue Proteins/genetics
17.
Int J Mol Sci ; 24(9)2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37175614

ABSTRACT

Despite significant advances in targeted therapies against the hyperactivated BRAFV600/MEK pathway for patients with unresectable metastatic melanoma, acquired resistance remains an unsolved clinical problem. In this study, we focused on melanoma cells resistant to trametinib, an agent broadly used in combination therapies. Molecular and cellular changes were assessed during alternating periods of trametinib withdrawal and rechallenge in trametinib-resistant cell lines displaying either a differentiation phenotype (MITFhigh/NGFRlow) or neural crest stem-like dedifferentiation phenotype (NGFRhigh/MITFlow). Neither drug withdrawal nor drug rechallenge induced cell death, and instead of loss of fitness, trametinib-resistant melanoma cells adapted to altered conditions by phenotype switching. In resistant cells displaying a differentiation phenotype, trametinib withdrawal markedly decreased MITF level and activity, which was associated with reduced cell proliferation capacity, and induced stemness assessed as NGFR-positive cells and senescence features, including IL-8 expression and secretion. All these changes could be reversed by trametinib re-exposure, which emphasizes melanoma cell plasticity. Trametinib-resistant cells displaying a dedifferentiation phenotype were less responsive presumably due to the already low level of MITF, a master regulator of the melanoma phenotype. Considering new directions of the development of anti-melanoma treatment, our study suggests that the phenotype of melanomas resistant to targeted therapy might be a crucial determinant of the selection of second-line therapy for melanoma patients.


Subject(s)
Interleukin-8 , Melanoma , Humans , Interleukin-8/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Phenotype , Proto-Oncogene Proteins B-raf/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Nerve Tissue Proteins/genetics , Receptors, Nerve Growth Factor/genetics
18.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047730

ABSTRACT

This study aimed to investigate the influence of genetic variants in neuroplasticity-related genes on antidepressant treatment phenotypes. The BDNF-TrkB signaling pathway, as well as the downstream kinases Akt and ERK and the mTOR pathway, have been implicated in depression and neuroplasticity. However, clinicians still struggle with the unpredictability of antidepressant responses in depressed patients. We genotyped 26 polymorphisms in BDNF, NTRK2, NGFR, CREB1, GSK3B, AKT, MAPK1, MTOR, PTEN, ARC, and SYN1 in 80 patients with major depressive disorder treated according to the Texas Medical Algorithm for 27 months at Hospital Magalhães Lemos, Porto, Portugal. Our results showed that BDNF rs6265, PTEN rs12569998, and SYN1 rs1142636 SNP were associated with treatment-resistant depression (TRD). Additionally, MAPK1 rs6928 and GSK3B rs6438552 gene polymorphisms were associated with relapse. Moreover, we found a link between the rs6928 MAPK1 polymorphism and time to relapse. These findings suggest that the BDNF, PTEN, and SYN1 genes may play a role in the development of TRD, while MAPK1 and GSK3B may be associated with relapse. GO analysis revealed enrichment in synaptic and trans-synaptic transmission pathways and glutamate receptor activity with TRD-associated genes. Genetic variants in these genes could potentially be incorporated into predictive models of antidepressant response.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Humans , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Nerve Tissue Proteins/genetics , Phenotype , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-akt/genetics , PTEN Phosphohydrolase/genetics , Receptors, Nerve Growth Factor/genetics , Recurrence , TOR Serine-Threonine Kinases/genetics
19.
Proc Natl Acad Sci U S A ; 120(6): e2205426120, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36730190

ABSTRACT

In neurons, many membrane proteins, synthesized in cell bodies, must be efficiently delivered to axons to influence neuronal connectivity, synaptic communication, and repair. Previously, we found that axonal targeting of TrkA neurotrophin receptors in sympathetic neurons occurs via an atypical transport mechanism called transcytosis, which relies on TrkA interactions with PTP1B, a protein tyrosine phosphatase. Here, we generated TrkAR685A mice, where TrkA receptor signaling is preserved, but its PTP1B-dependent transcytosis is disrupted to show that this mode of axonal transport is essential for sympathetic neuron development and autonomic function. TrkAR685A mice have decreased axonal TrkA levels in vivo, loss of sympathetic neurons, and reduced innervation of targets. The neuron loss and diminished target innervation phenotypes are specifically restricted to the developmental period when sympathetic neurons are known to rely on the TrkA ligand, nerve growth factor, for trophic support. Postnatal TrkAR685A mice exhibit reduced pupil size and eyelid ptosis, indicative of sympathetic dysfunction. Furthermore, we also observed a significant loss of TrkA-expressing nociceptive neurons in the dorsal root ganglia during development in TrkAR685A mice, suggesting that transcytosis might be a general mechanism for axonal targeting of TrkA receptors. Together, these findings establish the necessity of transcytosis in supplying TrkA receptors to axons, specifically during development, and highlight the physiological relevance of this axon targeting mechanism in the nervous system.


Subject(s)
Neurons , Receptor, trkA , Mice , Animals , Receptor, trkA/genetics , Receptor, trkA/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/genetics , Axons/metabolism , Transcytosis , Sympathetic Nervous System/metabolism
20.
Exp Neurol ; 359: 114161, 2023 01.
Article in English | MEDLINE | ID: mdl-35787888

ABSTRACT

The precursor form of nerve growth factor (proNGF) is essential to maintain NGF survival signaling. ProNGF is also among endogenous ligands for p75 neurotrophin receptor (p75ntr). Mounting evidence implies that p75ntr signaling contributes to neural damage in ischemic stroke. The present study examines the therapeutic effect of the p75ntr modulator LM11A-31. Adult mice underwent transient distal middle cerebral artery occlusion (t-dMCAO) followed by LM11A-31 treatment (25 mg/kg, i.p., twice daily) either for 72 h post-injury (acute phase) or afterward till two weeks post-stroke (subacute phase). LM11A-31 reduced blood-brain barrier permeability, cerebral tissue injury, and sensorimotor function in the acute phase of stroke. Ischemic brain samples showed repressed proNGF/P75ntr signaling and Caspase 3 activation in LM11A-31 treated mice, where we observed less reactive microglia and IL-1ß production. LM11A-31 (20-80 nM) also mitigated neural injury induced by oxygen-glucose deprivation (OGD) in sandwich co-cultures of primary cortical neurons (PCN) and astrocytes. This concurred with JNK/PARP downregulation and reduced caspase-3 cleavage in the PCNs and was associated with repressed proNGF generation in astrocytes. Further in vitro experiments indicated human proNGF suppresses the pro-inflammatory phenotype in microglial cultures, as determined by a sharp decline in HMGB-1 production and moderate arginase-1 upregulation. Despite significant protection in acute stroke, LM11A-31 treatment did not improve cortical atrophy and sensorimotor function in the subacute phase. Our findings provide preclinical evidence supporting LM11A-31 as a promising therapy for acute stroke injury. Further investigations may elucidate if reduced astrocytic proNGF, an endogenous reservoir of pro-neurotrophins, may restrict the therapeutic window.


Subject(s)
Receptor, Nerve Growth Factor , Stroke , Mice , Humans , Animals , Receptor, Nerve Growth Factor/metabolism , Nerve Growth Factor/metabolism , Astrocytes/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Stroke/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL