scholarly journals Novel Insights into the Cellular Localization and Regulation of the Autophagosomal Proteins LC3A, LC3B and LC3C

Cells ◽  
2020 ◽  
Vol 9 (10) ◽  
pp. 2315
Author(s):  
Marius W. Baeken ◽  
Katja Weckmann ◽  
Philip Diefenthäler ◽  
Jan Schulte ◽  
Kamran Yusifli ◽  
...  

Macroautophagy is a conserved degradative process for maintaining cellular homeostasis and plays a key role in aging and various human disorders. The microtubule-associated protein 1A/1B light chain 3B (MAP1LC3B or LC3B) is commonly analyzed as a key marker for autophagosomes and as a proxy for autophagic flux. Three paralogues of the LC3 gene exist in humans: LC3A, LC3B and LC3C. The molecular function, regulation and cellular localization of LC3A and LC3C have not been investigated frequently, even if a similar function to that described for LC3B appears likely. Here, we have selectively decapacitated LC3B by three separate strategies in primary human fibroblasts and analyzed the evoked effects on LC3A, LC3B and LC3C in terms of their cellular distribution and co-localization with p62, a ubiquitin and autophagy receptor. First, treatment with pharmacological sirtuin 1 (SIRT1) inhibitors to prevent the translocation of LC3B from the nucleus into the cytosol induced an increase in cytosolic LC3C, a heightened co-localization of LC3C with p62, and an increase LC3C-dependent autophagic flux as assessed by protein lipidation. Cytosolic LC3A, however, was moderately reduced, but also more co-localized with p62. Second, siRNA-based knock-down of SIRT1 broadly reproduced these findings and increased the co-localization of LC3A and particularly LC3C with p62 in presumed autophagosomes. These effects resembled the effects of pharmacological sirtuin inhibition under normal and starvation conditions. Third, siRNA-based knock-down of total LC3B in cytosol and nucleus also induced a redistribution of LC3C as if to replace LC3B in the nucleus, but only moderately affected LC3A. Total protein expression of LC3A, LC3B, LC3C, GABARAP and GABARAP-L1 following LC3B decapacitation was unaltered. Our data indicate that nuclear trapping and other causes of LC3B functional loss in the cytosol are buffered by LC3A and actively compensated by LC3C, but not by GABARAPs. The biological relevance of the potential functional compensation of LC3B decapacitation by LC3C and LC3A warrants further study.

2021 ◽  
Vol 15 ◽  
Author(s):  
Omid Vakili ◽  
Seyyed Hossein Khatami ◽  
Amir Maleksabet ◽  
Ahmad Movahedpour ◽  
Saeed Ebrahimi Fana ◽  
...  

Aims: Bioinformatics analysis of suitable signal peptide for recombinant glucarpidase. Background: Methotrexate (MTX) is a general chemotherapeutic agent utilized to treat a variety of malignancies., woefully, its high doses can cause nephrotoxicity and subsequent defect in the process of MTX excretion. The recombinant form of glucarpidase, is produced by engineered E. coli and is a confirmed choice to overcoming this problem. Objective: In the present study, in silico analyses were performed to select suitable SPs for the secretion of recombinant glucarpidase in E. coli. Methods: The signal peptide website and UniProt database were employed to collect the SPs and protein sequences. In the next step, SignalP-5.0 helped us to predict the SPs and the position of cleavage sites. Moreover, physicochemical properties and solubility were evaluated using ProtParam and Protein-sol online software, and finally, ProtCompB was used to predict the final sub-cellular localization. Results: Luckily, all SPs could form soluble fusion proteins. At last, it was found that PPB and TIBA could translocate the glucarpidase into the extracellular compartment. Conclusion: This study showed that there are only 2 applicable SPs for the extracellular translocation of glucarpidase. Although the findings were remarkable with high degrees of accuracy and precision based on the utilization of bioinformatics analyses, additional experimental assessments are required to confirm and validate it. Recent patents revealed several inventions related to the clinical aspects of vaccine peptide against human disorders.


2021 ◽  
Vol 11 ◽  
Author(s):  
Jane L. Roberts ◽  
Laurence Booth ◽  
Andrew Poklepovic ◽  
Paul Dent

We have extended our analyses of HDAC inhibitor biology in sarcoma. The multi-kinase inhibitor axitinib interacted with multiple HDAC inhibitors to kill sarcoma cells. Axitinib and HDAC inhibitors interacted in a greater than additive fashion to inactivate AKT, mTORC1 and mTORC2, and to increase Raptor S722/S792 phosphorylation. Individually, all drugs increased phosphorylation of ATM S1981, AMPKα T172, ULK1 S317 and ATG13 S318 and reduced ULK1 S757 phosphorylation; this correlated with enhanced autophagic flux. Increased phosphorylation of ULK1 S317 and of Raptor S722/S792 required ATM-AMPK signaling. ULK1 S757 is a recognized site for mTORC1 and knock down of either ATM or AMPKα reduced the drug-induced dephosphorylation of this site. Combined exposure of cells to axitinib and an HDAC inhibitor significantly reduced the expression of HDAC1, HDAC2, HDAC3, HDAC4, HDAC6 and HDAC7. No response was observed for HDACs 10 and 11. Knock down of ULK1, Beclin1 or ATG5 prevented the decline in HDAC expression, as did expression of a constitutively active mTOR protein. Axitinib combined with HDAC inhibitors enhanced expression of Class I MHCA and reduced expression of PD-L1 which was recapitulated via knock down studies, particularly of HDACs 1 and 3. In vivo, axitinib and the HDAC inhibitor entinostat interacted to significantly reduce tumor growth. Collectively our findings support the exploration of axitinib and HDAC inhibitors being developed as a novel sarcoma therapy.


2015 ◽  
Vol 22 (10) ◽  
pp. 1714-1726 ◽  
Author(s):  
M Mrschtik ◽  
J O'Prey ◽  
L Y Lao ◽  
J S Long ◽  
F Beaumatin ◽  
...  

Abstract Macroautophagy is a membrane-trafficking process that delivers cytoplasmic constituents to lysosomes for degradation. The process operates under basal conditions as a mechanism to turnover damaged or misfolded proteins and organelles. As a result, it has a major role in preserving cellular integrity and viability. In addition to this basal function, macroautophagy can also be modulated in response to various forms of cellular stress, and the rate and cargoes of macroautophagy can be tailored to facilitate appropriate cellular responses in particular situations. The macroautophagy machinery is regulated by a group of evolutionarily conserved autophagy-related (ATG) proteins and by several other autophagy regulators, which either have tissue-restricted expression or operate in specific contexts. We report here the characterization of a novel autophagy regulator that we have termed DRAM-3 due to its significant homology to damage-regulated autophagy modulator (DRAM-1). DRAM-3 is expressed in a broad spectrum of normal tissues and tumor cells, but different from DRAM-1, DRAM-3 is not induced by p53 or DNA-damaging agents. Immunofluorescence studies revealed that DRAM-3 localizes to lysosomes/autolysosomes, endosomes and the plasma membrane, but not the endoplasmic reticulum, phagophores, autophagosomes or Golgi, indicating significant overlap with DRAM-1 localization and with organelles associated with macroautophagy. In this regard, we further proceed to show that DRAM-3 expression causes accumulation of autophagosomes under basal conditions and enhances autophagic flux. Reciprocally, CRISPR/Cas9-mediated disruption of DRAM-3 impairs autophagic flux confirming that DRAM-3 is a modulator of macroautophagy. As macroautophagy can be cytoprotective under starvation conditions, we also tested whether DRAM-3 could promote survival on nutrient deprivation. This revealed that DRAM-3 can repress cell death and promote long-term clonogenic survival of cells grown in the absence of glucose. Interestingly, however, this effect is macroautophagy-independent. In summary, these findings constitute the primary characterization of DRAM-3 as a modulator of both macroautophagy and cell survival under starvation conditions.


Circulation ◽  
2020 ◽  
Vol 141 (25) ◽  
pp. 2095-2105 ◽  
Author(s):  
Milton Packer

Sodium-glucose cotransporter 2 inhibitors reduce the risk of serious heart failure and adverse renal events, but the mechanisms that underlie this benefit are not understood. Treatment with SGLT2 inhibitors is distinguished by 2 intriguing features: ketogenesis and erythrocytosis. Both reflect the induction of a fasting-like and hypoxia-like transcriptional paradigm that is capable of restoring and maintaining cellular homeostasis and survival. In the face of perceived nutrient and oxygen deprivation, cells activate low-energy sensors, which include sirtuin-1 (SIRT1), AMP-activated protein kinase (AMPK), and hypoxia inducible factors (HIFs; especially HIF-2α); these enzymes and transcription factors are master regulators of hundreds of genes and proteins that maintain cellular homeostasis. The activation of SIRT1 (through its effects to promote gluconeogenesis and fatty acid oxidation) drives ketogenesis, and working in concert with AMPK, it can directly inhibit inflammasome activation and maintain mitochondrial capacity and stability. HIFs act to promote oxygen delivery (by stimulating erythropoietin and erythrocytosis) and decrease oxygen consumption. The activation of SIRT1, AMPK, and HIF-2α enhances autophagy, a lysosome-dependent degradative pathway that removes dangerous constituents, particularly damaged mitochondria and peroxisomes, which are major sources of oxidative stress and triggers of cellular dysfunction and death. SIRT1 and AMPK also act on sodium transport mechanisms to reduce intracellular sodium concentrations. It is interesting that type 2 diabetes mellitus, obesity, chronic heart failure, and chronic kidney failure are characterized by the accumulation of intracellular glucose and lipid intermediates that are perceived by cells as indicators of energy overabundance. The cells respond by downregulating SIRT1, AMPK, and HIF-2α, thus leading to an impairment of autophagic flux and acceleration of cardiomyopathy and nephropathy. SGLT2 inhibitors reverse this maladaptive signaling by triggering a state of fasting and hypoxia mimicry, which includes activation of SIRT1, AMPK, and HIF-2α, enhanced autophagic flux, reduced cellular stress, decreased sodium influx into cells, and restoration of mitochondrial homeostasis. This mechanistic framework clarifies the findings of large-scale randomized trials and the close association of ketogenesis and erythrocytosis with the cardioprotective and renoprotective benefits of these drugs.


2003 ◽  
Vol 278 (40) ◽  
pp. 38966-38969 ◽  
Author(s):  
Gil Blander ◽  
Rita Machado de Oliveira ◽  
Caitlin M. Conboy ◽  
Marcia Haigis ◽  
Leonard Guarente

2020 ◽  
Author(s):  
Li Jiang ◽  
Yixin Zhang ◽  
Feng Jing ◽  
Ting Long ◽  
Guangcheng Qin ◽  
...  

Abstract Background: Central sensitization is an important pathophysiological mechanism of chronic migraine (CM). According to our previous studies, microglial activation and subsequent inflammation in the trigeminal nucleus caudalis (TNC) contribute to the central sensitization. The P2X7 receptor (P2X7R) is a purinergic receptor expressed in microglia and participates in central sensitization in chronic pain, but its role in CM is unclear. Numerous studies have shown that P2X7R regulates the level of autophagy and that autophagy affects the microglial activation and inflammation. Recently, autophagy has been shown to be involved in neuropathic pain, but there is no information about autophagy in CM. Therefore, the current study investigated the role of P2X7R in CM and its underlying mechanism, focusing on autophagy regulation.Methods: The CM model was established by repeated intraperitoneal injection of nitroglycerin (NTG) in mice. A Von Frey filament and radiant heat were used to assess the mechanical and thermal hypersensitivity. Western blotting and immunofluorescence assays were performed to detect the expression of P2X7R, autophagy-related proteins, and the cellular localization of P2X7R. To determine the role of P2X7R and autophagy in CM, we detected the effects of the autophagy inducer, rapamycin (RAPA) and P2X7R antagonist, Brilliant Blue G (BBG), on pain behavior and the expression of calcitonin gene-related peptide (CGRP) and c-fos. In addition, the effect of RAPA and BBG on microglial activation and subsequent inflammation were investigated.Results: The expression of P2X7R was increased and was mainly colocalized with microglia in the TNC following recurrent NTG administration. The autophagic flux was blocked in CM, which was characterized by up-regulated LC3-II, and accumulated autophagy substrate protein, p62. RAPA significantly improved the basal rather than acute hyperalgesia. BBG alleviated both basal and acute hyperalgesia. BBG activated the level of autophagic flux. RAPA and BBG inhibited the activation of microglia, limited the inflammatory response, and reduced the expression of CGRP and c-fos. Conclusions: Our results demonstrate the dysfunction of the autophagic process in CM. Activated autophagy may have a preventive effect on migraine chronification. P2X7R contributes to central sensitization through mediating autophagy regulation and might become a potential target for CM.


2019 ◽  
Vol 316 (5) ◽  
pp. C605-C620 ◽  
Author(s):  
Nadia Bouhamdani ◽  
Dominique Comeau ◽  
Kevin Cormier ◽  
Sandra Turcotte

Autophagy is a highly conserved, homeostatic process by which cytosolic components reach lysosomes for degradation. The roles played by different autophagic processes in cancer are complex and remain cancer type and stage dependent. Renal cell carcinoma (RCC) is the most common subtype of kidney cancer and is characterized by the inactivation of the von Hippel-Lindau (VHL) tumor suppressor. Our previous study identified a small compound, STF-62247, as an autophagy-modulating molecule causing selective cytotoxicity for VHL-inactivated cells. This present study investigates the effects of STF-62247 specifically on the macroautophagic flux to better characterize its mechanism of action in RCC. Our results clearly demonstrate that this compound is a potent blocker of late stages of autophagy. We show that inhibiting autophagy by CRISPR knockouts of autophagy-related genes rendered VHL-deficient cells insensitive to STF-62247, uncovering the importance of the autophagic pathway in STF-selective cell death. By exploiting the autofluorescence of STF-62247, we pinpointed its cellular localization to lysosomes. Finally, in response to prolonged STF treatments, we show that VHL-proficient cells are able to surmount the block in late stages of autophagy by restoring their lysosome numbers. Conversely, an increase in autophagic vesicles accompanied by a time-dependent decrease in lysosomes was observed in VHL-deficient cells. This is the first mechanistic study investigating STF-62447’s effects on the autophagic flux in RCC. Importantly, our study reclassifies STF-62247 as a blocker of later stages of autophagy and highlights the possibility of blocking this process through lysosome disruption in VHL-mutated RCCs.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2216-2216
Author(s):  
Sophia E. Maharry ◽  
Sujay Mehta ◽  
Sandya Liyanarachchi ◽  
Christopher J. Walker ◽  
Clara D. Bloomfield ◽  
...  

Abstract Background: Overcoming the block of differentiation is a major effort in Acute Myeloid Leukemia (AML) research. We visited the major quantitative trait locus (QTL) for hematopoietic differentiation on chromosome 6q23 to identify genes involved in hematopoietic development that are possibly deregulated during leukemogenesis. We noticed a novel microRNA within the QTL: miR-3662. Aims: The goal was to elucidate a possible causal involvement of miR-3662 in the association of the 6q23 QTL with hematopoietic differentiation and/or leukemogenesis. Results: Monitoring of miR-3662 expression during cytokine stimulated differentiation of CD34+ hematopoietic progenitor cells (HPCs) revealed increasing miR-3662 abundance during differentiation, with the highest expression found in terminally differentiated cells. In contrast, miR-3662 had very low expression in AML cell lines (KG1a, MV4-11, OCI-AML3, THP1) and primary AML patient blasts (n=12). We stably introduced miR-3662 into HPCs of 3 non-leukemic donors, and differentiated the cells into erythroid, megakaryocytic and granulocytic lineages. Forced expression of miR-3662 led to significant increase of colony formation, most pronounced in the erythroid lineage. In contrast, targeted knock-down of miR-3662 significantly reduced colony formation (Figure 1). To test if the observed downregulation of miR-3662 in leukemic cells is favorable to leukemic cells, we stably introduced miR-3662 in AML cell lines (MV4-11, KG1a) and AML patient blasts (n=12). Increasing the abundance of miR-3662 led to reduction of cell viability, increased cell death and reduced colony formation. To find the downstream targets of miR-3662, we performed a comprehensive targeted RNA sequencing approach with a panel of 361 genes (TruSeq platform, Illumina; followed by Ingenuity Pathway analysis), using RNA from two AML patient blasts infected with miR-3662 vs. scramble. miR-3662 was associated with >20% reduction in the mRNA expression of 34 genes. Among these, 6 genes harbored predicted miR-3662 binding sites in their 3′-UTRs, qualifying as potential direct miR-3662 targets. Of these, the inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB) had the highest affinity score for miR-3662, and was thus chosen as the primary candidate. Determination of endogenous miR-3662 and IKBKB expression in primary AML patient blasts (n=12) showed a significant association of higher miR-3662 with lower IKBKB abundance. qPCR and Western blotting confirmed miR-3662 mediated downregulation of IKBKB in HPCs, KG1a and MV4-11 cells and primary patient blasts. Because IKBKB leads to reduced phosphorylation of the inhibitor in the inhibitor/NF-ĸB complex (IKBa), it limits NF-ĸB's nuclear localization and consequent transcriptional activation. Increasing miR-3662 should thus decrease NF-ĸB's nuclear localization. Western blotting showed reduced phosphorylation of IKBa and confocal imaging showed less NF-ĸB in the nucleus in miR-3662 expressing MV4-11 and HPC cells (Figure 2). Luciferase assay validated a direct interaction of miR-3662 with the 3′-UTR of IKBKB. Finally, we attempted to gain insights into the upstream regulation of miR-3662. We identified a possible transcription start site for miR-3662 (TSS-3662), whose activation potential was validated by both luciferase assays and enrichment of RNA polymerase II (Pol II), histone H3 methylated Lys (H3K4), and total histone H3 CHIP assays of TSS-3662. Testing of predicted activating transcription factors showed GATA1 and CEBPA to have an activating potential (as shown by luciferase assays) and binding affinity (as shown by electrophoretic mobility shift assays) to TSS-3662. Overexpression of GATA1 and CEBPA significantly increased miR-3662's abundance. Conclusion: We have identified miR-3662, located in the major hematopoietic QTL on chromosome 6q23, as a novel regulator of hematopoietic differentiation that acts by regulating the NF-ĸB pathway. Reduced abundance of miR-3662 in leukemic cells potentially contributes to the de-differentiated phenotype of leukemic cells and to increased cell growth by increasing nuclear localization of NF-ĸB. Figure 1. Effect of miR-3662 overexpression and knock-down on colony formation of HPCs. Figure 1. Effect of miR-3662 overexpression and knock-down on colony formation of HPCs. Figure 2. Cellular localization of NFĸB in miR-3662 vs. scramble-infected HPCs. Figure 2. Cellular localization of NFĸB in miR-3662 vs. scramble-infected HPCs. Disclosures Walker: Karyopharm Therapeutics Inc.: Research Funding.


2017 ◽  
Vol 2017 ◽  
pp. 1-16 ◽  
Author(s):  
Ana Ortiz-Espín ◽  
Esther Morel ◽  
Ángeles Juarranz ◽  
Antonio Guerrero ◽  
Salvador González ◽  
...  

The Antarctic plantDeschampsia antarctica(DA) is able to survive in extreme conditions thanks to its special mechanism of protection against environmental aggressions. In this work, we investigated whether an aqueous extract of the plant (EDA) retains some of its defensive properties and is able to protect our skin against common external oxidants. We evaluated EDA over young human fibroblasts and exposed to H2O2, and we measured cell proliferation, viability, and senescence-associatedβ-galactosidase (SA-β-Gal). We also tested the expression of several senescence-associated proteins including sirtuin1, lamin A/C, the replicative protein PCNA, and the redox protein thioredoxin 2. We found that EDA promotedper secell proliferation and viability and increased the expression of anti-senescence-related markers. Then, we selected a dose of H2O2as an inductor of senescence in human fibroblasts, and we found that an EDA treatment 24 h prior H2O2exposure increased fibroblast proliferation. EDA significantly inhibited the increase in SA-β-Gal levels induced by H2O2and promoted the expression of sirtuin 1 and lamin A/C proteins. Altogether, these results suggest that EDA protects human fibroblasts from cellular senescence induced by H2O2, pointing to this compound as a potential therapeutic agent to treat or prevent skin senescence.


Biomolecules ◽  
2020 ◽  
Vol 10 (5) ◽  
pp. 761
Author(s):  
Seon Beom Song ◽  
Eun Seong Hwang

Under glucose deprivation, cells heavily mobilize oxidative phosphorylation to maintain energy homeostasis. This leads to the generation of high levels of ATP, as well as reactive oxygen species (ROS), from mitochondria. In nutrient starvation, autophagy is activated, likely to facilitate resource recycling, but recent studies suggest that autophagy flux is inhibited in cells undergoing glucose deprivation. In this study, we analyzed the status of autophagic flux in glucose-deprived human fibroblasts. Although lysosomes increased in quantity due in part to an increase of biogenesis, a large population of them suffered low acidity in the glucose-deprived cells. Autophagosomes also accumulated due to poor autolysis in these cells. A treatment of antioxidants not only restored lysosomal acidity but also released the flux blockade. The inhibition of ataxia telangiectasia mutated (ATM) serine/threonine kinase, which is activated by ROS, also attenuated the impairment of lysosomal acidity and autophagic flux, suggesting an effect of ROS that might be mediated through ATM activation. In addition, the activity of extracellular signal-regulated kinase (Erk) increased upon glucose deprivation, but this was also compromised by a treatment of antioxidants. Furthermore, the Erk inhibitor treatment also alleviated the failure in lysosomal acidity and autophagic flux. These together indicate that, upon glucose deprivation, cells undergo a failure of autophagy flux through an impairment of lysosomal acidity and that a high-level ROS-induced activation of Erk and ATM is involved in this impairment.


Sign in / Sign up

Export Citation Format

Share Document