aldolase a
Recently Published Documents


TOTAL DOCUMENTS

220
(FIVE YEARS 31)

H-INDEX

29
(FIVE YEARS 3)

2021 ◽  
Author(s):  
Kathryn D. Fenton ◽  
Kathleen M. Meneely ◽  
Tiffany Wu ◽  
Tyler A. Martin ◽  
Liskin Swint‐Kruse ◽  
...  

2021 ◽  
Author(s):  
Shahan Mamoor

We mined published microarray data (1) to understand the most significant gene expression differences in the tumors of triple negative breast cancer patients based on survival following treatment: dead or alive. We observed significant transcriptome-wide differential expression of aldolase A, fructose-bisphosphate, encoded by ALDOA when comparing the primary tumors of triple negative breast cancer patients dead or alive. Importantly, ALDOA expression was correlated with overall survival in basal subtype breast cancer, a molecular subtype sharing significant overlap with triple negative breast cancer. ALDOA may be of relevance as a biomarker or as a molecule of interest in understanding the etiology or progression of triple negative breast cancer.


2021 ◽  
Vol 16 (3) ◽  
pp. 380-386
Author(s):  
Iulian Constantin CREANGĂ ◽  
◽  
Alexandra RIPSZKY TOTAN ◽  
Daniela MIRICESCU ◽  
Iulia-Ioana STĂNESCU-SPÎNU ◽  
...  

Introduction. Crushing syndrome can affect all structures of a limb and can cause unique or complex injuries. The clinical picture of the injuries is nonspecific and extremely varied, and is not characteristic of the mechanism that generated it. The aim of our study was to correlate the changes of ALD A (aldolase A) and CAIII (carbonic anhydrase III) in the appearance and evolution of the crushing syndrome. Material and methods. The present study included 22 patients with crushing syndrome. The control group consisted of 20 healthy subjects. IL-6 (interleukin-6), CK (creatine kinase), ALD A and CAIII blood levels were determined simultaneously from all the participants to the study. Statistical analysis was performed using IBM SPSS Statistics 25 and Microsoft Office Excel / Word 2013. Results. Our results showed significantly increased levels of ALD A, CAIII, IL-6 and CK in patients with crushing syndrome. However, no statistically significant correlations could be found between the studied biomarkers. Discussions. So far, in the medical literature, very few studies focused on the correlation between the occurrence of crushing syndrome and changes in ALD A and CA III levels. Conclusions. Our study showed that ALD A and CAIII may become important biomarkers for the rapid diagnosis of crushing syndrome in the future.


2021 ◽  
Vol 17 (9) ◽  
pp. e1009943
Author(s):  
Saray Gutiérrez ◽  
Julia Fischer ◽  
Raja Ganesan ◽  
Nina Judith Hos ◽  
Gökhan Cildir ◽  
...  

Regulation of cellular metabolism is now recognized as a crucial mechanism for the activation of innate and adaptive immune cells upon diverse extracellular stimuli. Macrophages, for instance, increase glycolysis upon stimulation with pathogen-associated molecular patterns (PAMPs). Conceivably, pathogens also counteract these metabolic changes for their own survival in the host. Despite this dynamic interplay in host-pathogen interactions, the role of immunometabolism in the context of intracellular bacterial infections is still unclear. Here, employing unbiased metabolomic and transcriptomic approaches, we investigated the role of metabolic adaptations of macrophages upon Salmonella enterica serovar Typhimurium (S. Typhimurium) infections. Importantly, our results suggest that S. Typhimurium abrogates glycolysis and its modulators such as insulin-signaling to impair macrophage defense. Mechanistically, glycolysis facilitates glycolytic enzyme aldolase A mediated v-ATPase assembly and the acidification of phagosomes which is critical for lysosomal degradation. Thus, impairment in the glycolytic machinery eventually leads to decreased bacterial clearance and antigen presentation in murine macrophages (BMDM). Collectively, our results highlight a vital molecular link between metabolic adaptation and phagosome maturation in macrophages, which is targeted by S. Typhimurium to evade cell-autonomous defense.


2021 ◽  
Author(s):  
Ya Lu ◽  
Yuan Zhang ◽  
Hui Zhang ◽  
Yue Zhu ◽  
Junying Zhang ◽  
...  

Abstract Background: Colorectal cancer (CRC) is a serious threat to human health, and its underlying mechanisms needs further explored. Aldolase A (ALDOA) has received increasing attention for its reported association with multiple cancers, but the function and mechanism of ALDOA in CRC remain unclear. We aimed to evaluate the biological role of ALDOA in CRC.Methods: The stable ALDOA knockdown or overexpression cell lines were established for subsequent experiments. The qRT-PCR and western blotting were used to detect the expression of ALDOA and COPS6 and the relative protein levels of epithelial-mesenchymal transition (EMT) and MAPK signaling pathway. Immunofluorescence (IF) assay was applied to determine ALDOA localization. CCK-8, transwell, and wound healing assays were performed to evaluate CRC cell proliferation, invasion, and migration. Mouse xenograft models were established to verify the effect of ALDOA on CRC cell growth in vivo. Immunoprecipitation (IP) assay and mass spectrometry (MS) analysis were conducted to identify the interactions between ALDOA and COPS6.Results: ALDOA was overexpressed in CRC tissues and cell lines. Silencing ALDOA significantly impaired the proliferation, invasion and migration of CRC cells in vitro, and obviously decreased the growth of CRC cells in vivo. Mechanically, ALDOA bound to and regulated COPS6, and the promoting effects of upregulated ALDOA on CRC cell proliferation and metastasis were inhibited by the depletion of COPS6. Besides, EMT program and MAPK signaling pathway were activated by ALDOA overexpression.Conclusion: ALDOA facilitated the proliferation, invasion and migration of CRC through binding and regulating COPS6, inducing EMT and activating MAPK signaling pathway.


Author(s):  
Esra Kara ◽  
Deniz Kor ◽  
Fatma Derya Bulut ◽  
Özlem Hergüner ◽  
Serdar Ceylaner ◽  
...  

Abstract Objectives Aldolase A deficiency also known as glycogen storage disease (GSD) XII, is an ultra rare autosomal recessively inherited GSD, associated with hemolytic anemia and rhabdomyolysis. Case presentation Here, we first report a patient with dermatological findings, hemodialysis requirement for rhabdomyolysis, and a novel likely pathogenic c.971C>T (p.A324V) mutation in the ALDOA gene. Conclusions Episodes of rhabdomyolysis can be triggered by febrile illnesses and catabolic processes. Diagnosis should be confirmed by the mutation analysis of ALDOA gene. Treatment includes management of hemolytic anemia and administration of antipyretics during febrile episodes to avoid hemolysis and rhabdomyolysis.


2021 ◽  
Vol 27 ◽  
pp. 100730
Author(s):  
C. Papadopoulos ◽  
M. Svingou ◽  
K. Kekou ◽  
S. Vergnaud ◽  
S. Xirou ◽  
...  
Keyword(s):  

2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Duan Li ◽  
Xiaoyan Wang ◽  
Guangyu Li ◽  
Yujie Dang ◽  
Shidou Zhao ◽  
...  

AbstractGranulosa cell (GC) is a critical somatic component of ovarian follicles to support oocyte development, while the regulatory role of long noncoding RNA (lncRNA) in GCs is largely unknown. Here, we identified a down-regulated lncRNA ZNF674-AS1 in GCs from patients with biochemical premature ovarian insufficiency (bPOI), and its expression correlates with serum levels of clinical ovarian reserve indicators. Functional experiments showed that ZNF674-AS1 is induced by energy stress, and regulates the proliferation and glycolysis of GCs, which possibly leads to follicular dysfunction. Mechanistically, low-expressed ZNF674-AS1 reduced the enzymatic activity of aldolase A (ALDOA), concomitant with promoting the association between ALDOA and v-ATPase to activate the lysosome localized AMP-activated protein kinase (AMPK). These findings identified a new lncRNA–ALDOA complex through which ZNF674-AS1 exerts its functions, expanding the understanding of epigenetic regulation of GCs function and POI pathogenesis.


2021 ◽  
Author(s):  
Benjamin D Stein ◽  
Jun X Huang ◽  
David Wu ◽  
Lewis C Cantley ◽  
Raymond E Moellering

Hotspot thermal profiling (HTP) methods utilize modified-peptide level information in order to interrogate proteoform-specific stability inside of live cells. The first demonstration of HTP involved the integration of phosphopeptide enrichment into a TMT-based, single-LC separation thermal profiling workflow1. Here we present a new "label-fractionate-enrich" (LFE)-HTP method that involves high-pH reverse phase fractionation of TMT-labeled peptides prior to phosphopeptide enrichment, followed by peptide detection and quantitation using multi-notch LC-MS3. We find that LFE-HTP, while more resource intensive, improves the depth and precision of (phospho)proteoform coverage relative to the initial published HTP workflow. The fraction of detected phosphorylation sites that are significantly perturbed in this new dataset are consistent with those seen in our previous study, as well as those published by others, when compared head-to-head with the same analysis pipelines. Likewise, many hotspot phosphorylation sites identified in our paper are consistently reproduced by LFE-HTP as well as other modified HTP methods. The LFE-HTP dataset contains many novel hotspot phosphorylation sites that regulate the stability of diverse proteins, including phosphosites in the central glycolytic enzyme Aldolase A that are associated with monomer-to-oligomer formation, enzymatic activity and metabolic regulation in cancer cells. Our comparative analyses confirm that several variants of the HTP method can track modified proteoforms in live cells to detect and prioritize PTM-dependent changes in protein stability that may be associated with function.


Sign in / Sign up

Export Citation Format

Share Document