pathway dysregulation
Recently Published Documents


TOTAL DOCUMENTS

92
(FIVE YEARS 25)

H-INDEX

17
(FIVE YEARS 4)

2021 ◽  
pp. 1-10
Author(s):  
Suma P. Shankar ◽  
Reshmitha Fallurin ◽  
Tonya Watson ◽  
Prabhu R. Shankar ◽  
Terri L. Young ◽  
...  

2021 ◽  
Author(s):  
Weiping Tao ◽  
Xuesong Zhang ◽  
Juan Ding ◽  
Shijian Yu ◽  
Peiqing Ge ◽  
...  

Abstract Background: Brain-derived neurotrophic factor/tyrosine kinase receptor B (BDNF/TrkB) pathway dysregulation may be induced by hypoxia and inflammation, and play pivotal roles during the development of neurological disorders. Propofol is an anesthetic agent with neuro-protective properties. We aimed to verify whether propofol affected BDNF/TrkB pathway in neurons and astrocytes exposed to hypoxia and inflammation.Methods: Primary rat hippocampal neurons and astrocytes were cultured and exposed to propofol followed by hypoxia or TNF-α treatment. The production of BDNF and the expression/truncation/phosphorylation of TrkB were measured. The underlying mechanisms such as ERK, CREB, p35 and Cdk5 were investigated.Results: In hippocampal neurons and astrocytes, hypoxia and TNF-α reduced the production of BDNF. Pretreatment of hippocampal neurons with 25μM propofol reversed the inhibitory effect of hypoxia or TNF-α on BDNF production. However, even 100μM propofol had no such effect in astrocytes. Further, we found that in hippocampal neurons hypoxia and TNF-α increased the phosphorylation of ERK (p-ERK) and CREB at Ser142 (p-CREB Ser142), while reduced the phosphorylation of CREB at Ser133 (p-CREB Ser133), which were all reversed by 25μM propofol and 10μM ERK inhibitor. In addition, we reported that hypoxia- and TNF-α-mediated reduction of BDNF was mitigated by 10μM ERK inhibitor, and the beneficial effect of propofol was abolished by 10μM ERK activator. We also found neither hypoxia nor TNF-α affected TrkB expression, truncation or phosphorylation in hippocampal neurons and astrocytes. However 50μM propofol induced TrkB phosphorylation without affecting its expression and truncation only in hippocampal neurons. Furthermore, we detected that in hippocampal neurons, 50μM propofol induced p35 expression and Cdk5 activation, and blockade of p35 or Cdk5 mitigated propofol-induced TrkB phosphorylation.Conclusions: Propofol, via ERK/CREB and p35/Cdk5, may modulate BDNF/TrkB pathway in hippocampal neurons that were exposed to hypoxia or TNF-α.


2021 ◽  
Author(s):  
Chengnan Guo ◽  
Depeng Jiang ◽  
Yixi Xu ◽  
Fang Peng ◽  
Shuzhen Zhao ◽  
...  

Abstract Background Diabetic retinopathy (DR) is a major diabetes-related disease linked to metabolism. However, scientifically assessment of serum metabolic alterations in DR is scarce. We aimed to investigate the changes in metabolic coregulation from type 2 diabetic patients (T2DM) to DR and identify corresponding metabolite predictors via a widely targeted metabolomics approach.Methods In this case-control study, we tested 613 serum metabolites in 69 pairs of T2DM with DR (case) and propensity score-matched T2DM without DR (control) utilizing the ultra-performance liquid chromatography-electrospray ionization-tandem mass spectrometry system. The discrimination capability of differentially expressed metabolites (DEMs) in DR identification was also evaluated using a least absolute shrinkage and selection operator (LASSO) regression-based linear support vector machine (SVM) classifier. Metabolic pathway dysregulation in DR were comprehensively investigated by metabolic pathway analysis, chemical similarity enrichment analysis and MetaMapp approaches.Results A total of 89 DEMs were identified after paired univariate analysis and partial least squares discriminant analysis. The linear-SVM model based on LASSO regression selected DEMs had an excellent discrimination with an area under the ROC curve (AUC) as 0.99 (95% confidence interval: 0.95, 1.00). The biosynthesis of polyunsaturated fatty acids (PUFAs), thiamine metabolism, amino acids (mainly glycine, serine and threonine metabolism), hydroxyeicosatetraenoic acid (HETE), disaccharides, indoles and nucleotides were significantly enriched in DR. Conclusions This study systematically demonstrates that distinct metabolic alterations are linked to DR initiation. n-3 PUFAs, trehalose and vitamin B1 play an important role in inhibiting DR progression.


2021 ◽  
Author(s):  
Weiping Tao ◽  
Xuesong Zhang ◽  
Juan Ding ◽  
Shijian Yu ◽  
Peiqing Ge ◽  
...  

Abstract Background: BDNF/TrkB pathway dysregulation may be induced by hypoxia and inflammation, and play pivotal roles during the development of neurological disorders. Propofol is an anesthetic agent with neuro-protective properties. We aimed to verify whether propofol affected BDNF/TrkB pathway in neurons exposed to hypoxia or TNF-α.Methods: Primary rat hippocampal neurons and astrocytes were cultured and exposed to propofol followed by hypoxia or TNF-α treatment. The production of BDNF and the expression/truncation/phosphorylation of TrkB were measured. The underlying mechanisms such as ERK, CREB, p35 and Cdk5 were investigated.Results: In hippocampal neurons and astrocytes, hypoxia and TNF-α reduced the production of BDNF. Pretreatment of hippocampal neurons with 25μM propofol reversed the inhibitory effect of hypoxia or TNF-α on BDNF production. However, even 100μM propofol had no such effect in astrocytes. Further, we found that in hippocampal neurons hypoxia and TNF-α increased the phosphorylaion of ERK (p-ERK) and CREB at Ser142 (p-CREBSer142), while reduced the phosphorylation of CREB at Ser133 (p-CREBSer133), which were all reversed by 25μM propofol and 10μM ERK inhibitor. In addition, neither hypoxia nor TNF-α affected TrkB expression, truncation or phosphorylation in hippocampal neurons and astrocytes. However 50μM propofol induced TrkB phosphorylation without affecting its expression and truncation only in hippocampal neurons. Furthermore, we detected that in hippocampal neurons, 50μM propofol induced p35 expression and Cdk5 activation, and blockade of p35 or Cdk5 mitigated propofol-induced TrkB phosphorylation.Conclusions: Propofol, via ERK/CREB and p35/Cdk5, may modulate BDNF/TrkB pathway in hippocampal neurons that were exposed to hypoxia or TNF-α.


2021 ◽  
Vol 23 (Supplement_1) ◽  
pp. i44-i44
Author(s):  
Eliza Baird-Daniel ◽  
Shadi Zahedi ◽  
Andrew Morin ◽  
Michelle Desmarais ◽  
Kyle Williams ◽  
...  

Abstract Background Autophagy inhibition is a potential treatment for central nervous system (CNS) tumors. Autophagy, a heavily regulated process by which cellular waste is transferred to lysosomes for degradation and processing, is an integral part of tumor cell survival under stressful conditions including nutrient deprivation and chemotherapy. While the efficacy of autophagy inhibition has been demonstrated in CNS tumors with BRAFV600e mutations, it has yet to be explored in other CNS tumor types with MAPK pathway dysregulation including NF1-mutated tumors. Many tumors associated with the NF1 phenotype can be difficult to treat surgically thus development of further pharmacologic interventions is necessary. Methods A CRISPR/Cas9 mediated NF1 KO was derived from human immortalized Schwann cells and utilized as a tumor model. Autophagy inhibition was achieved pharmacologically by chloroquine (CQ) and genetically via shRNAi of ATG5 and ATG7. Trametinib was used for MEK inhibition. Cell growth and viability were determined by Incucyte, Cell Titer-Glo luminescent assay, and colony-formation assays. Protein expression was measured by western blot. Results We demonstrate increased autophagic activity in NF1 KO cell as compared to control lines both at baseline and in response to cellular stress. Furthermore, we describe that NF1 KO cells exhibit increased sensitivity to CQ alone, CQ in combination with trametinib, and shRNAi-mediated autophagy inhibition in combination with trametinib. Conclusion Here, we describe increased autophagic dependence of NF1 mutated tumors and demonstrate increased tumor sensitivity to autophagy inhibition both alone and in combination with MEK inhibition. These findings indicate that autophagy inhibition via CQ may be an effective adjunctive treatment for NF1 mutated tumors and suggests that diverse CNS tumor types with MAPK pathway dysregulation are susceptible to autophagy inhibition. Clinical investigation of combined MEK and autophagy inhibition has the potential to improve outcomes for NF1 patients with CNS tumors.


Oncogenesis ◽  
2021 ◽  
Vol 10 (5) ◽  
Author(s):  
Wen-Bin Ou ◽  
Meijun Z. Lundberg ◽  
Shuihao Zhu ◽  
Nacef Bahri ◽  
Anastasios Kyriazoglou ◽  
...  

AbstractEndometrial stromal sarcoma (ESS) is the second most common subtype of uterine mesenchymal cancer, after leiomyosarcoma, and oncogenic fusion proteins are found in many ESS. Our previous studies demonstrated transforming properties and diagnostic relevance of the fusion oncoprotein YWHAE–NUTM2 in high-grade endometrial stromal sarcoma (HG-ESS) and showed that cyclin D1 is a diagnostic biomarker in these HG-ESS. However, YWHAE–NUTM2 mechanisms of oncogenesis and roles in cyclin D1 expression have not been characterized. In the current studies, we show YWHAE-NUTM2 complexes with both BRAF/RAF1 and YAP/TAZ in HG-ESS. These interactions are functionally relevant because YWHAE-NUTM2 knockdown in HG-ESS and other models inhibits RAF/MEK/MAPK phosphorylation, cyclin D1 expression, and cell proliferation. Further, cyclin D1 knockdown in HG-ESS dephosphorylates RB1 and inhibits proliferation. In keeping with these findings, we show that MEK and CDK4/6 inhibitors have anti-proliferative effects in HG-ESS, and combinations of these inhibitors have synergistic activity. These findings establish that YWHAE-NUTM2 regulates cyclin D1 expression and cell proliferation by dysregulating RAF/MEK/MAPK and Hippo/YAP-TAZ signaling pathways. Recent studies demonstrate Hippo/YAP-TAZ pathway aberrations in many sarcomas, but this is among the first studies to demonstrate a well-defined oncogenic mechanism as the cause of Hippo pathway dysregulation.


CNS Oncology ◽  
2021 ◽  
pp. CNS68
Author(s):  
Timothy A Gregory ◽  
Lyndon B Chumbley ◽  
John W Henson ◽  
Brett J Theeler

Adult pilocytic astrocytoma (PA) is less prevalent than pediatric PA and is associated with a worse prognosis. In a literature review, we found that 88.3% of the molecular alterations in adult PA are associated with MAPK pathway dysregulation. The most common alterations are fusions of BRAF. Understanding of the mechanisms underlying this pathway has evolved substantially, heralding advancements in specific targeted therapy. Here, we review clinical and molecular features of adult PA, characteristics predicting aggressive behavior and approaches to standard and investigational therapies. We highlight epigenetic profiling and integrated diagnosis as an essential component of classifying PA.


Sign in / Sign up

Export Citation Format

Share Document