Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 153(5): 1064-79, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706743

ABSTRACT

Metabolic adaptation is essential for cell survival during nutrient deprivation. We report that eukaryotic elongation factor 2 kinase (eEF2K), which is activated by AMP-kinase (AMPK), confers cell survival under acute nutrient depletion by blocking translation elongation. Tumor cells exploit this pathway to adapt to nutrient deprivation by reactivating the AMPK-eEF2K axis. Adaptation of transformed cells to nutrient withdrawal is severely compromised in cells lacking eEF2K. Moreover, eEF2K knockdown restored sensitivity to acute nutrient deprivation in highly resistant human tumor cell lines. In vivo, overexpression of eEF2K rendered murine tumors remarkably resistant to caloric restriction. Expression of eEF2K strongly correlated with overall survival in human medulloblastoma and glioblastoma multiforme. Finally, C. elegans strains deficient in efk-1, the eEF2K ortholog, were severely compromised in their response to nutrient depletion. Our data highlight a conserved role for eEF2K in protecting cells from nutrient deprivation and in conferring tumor cell adaptation to metabolic stress. PAPERCLIP:


Subject(s)
Caenorhabditis elegans/metabolism , Elongation Factor 2 Kinase/metabolism , Neoplasms/physiopathology , Peptide Chain Elongation, Translational , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Animals , Brain Neoplasms/physiopathology , Caenorhabditis elegans/genetics , Cell Survival , Cell Transformation, Neoplastic , Elongation Factor 2 Kinase/genetics , Food Deprivation , Glioblastoma/physiopathology , HeLa Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Transplantation , Peptide Elongation Factor 2/metabolism , Transplantation, Heterologous
2.
EMBO Rep ; 24(12): e56815, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37846480

ABSTRACT

HACE1 is a HECT family E3 ubiquitin-protein ligase with broad but incompletely understood tumor suppressor activity. Here, we report a previously unrecognized link between HACE1 and signaling complexes containing mammalian target of rapamycin (mTOR). HACE1 blocks mTORC1 and mTORC2 activities by reducing mTOR stability in an E3 ligase-dependent manner. Mechanistically, HACE1 binds to and ubiquitylates Ras-related C3 botulinum toxin substrate 1 (RAC1) when RAC1 is associated with mTOR complexes, including at focal adhesions, leading to proteasomal degradation of RAC1. This in turn decreases the stability of mTOR to reduce mTORC1 and mTORC2 activity. HACE1 deficient cells show enhanced mTORC1/2 activity, which is reversed by chemical or genetic RAC1 inactivation but not in cells expressing the HACE1-insensitive mutant, RAC1K147R . In vivo, Rac1 deletion reverses enhanced mTOR expression in KRasG12D -driven lung tumors of Hace1-/- mice. HACE1 co-localizes with mTOR and RAC1, resulting in RAC1-dependent loss of mTOR protein stability. Together, our data demonstrate that HACE1 destabilizes mTOR by targeting RAC1 within mTOR-associated complexes, revealing a unique ubiquitin-dependent process to control the activity of mTOR signaling complexes.


Subject(s)
Ubiquitin-Protein Ligases , Animals , Mice , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , TOR Serine-Threonine Kinases , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
J Pathol ; 254(3): 254-264, 2021 07.
Article in English | MEDLINE | ID: mdl-33797756

ABSTRACT

Hereditary diffuse gastric cancer (HDGC) is a cancer syndrome caused by germline variants in CDH1, the gene encoding the cell-cell adhesion molecule E-cadherin. Loss of E-cadherin in cancer is associated with cellular dedifferentiation and poor prognosis, but the mechanisms through which CDH1 loss initiates HDGC are not known. Using single-cell RNA sequencing, we explored the transcriptional landscape of a murine organoid model of HDGC to characterize the impact of CDH1 loss in early tumourigenesis. Progenitor populations of stratified squamous and simple columnar epithelium, characteristic of the mouse stomach, showed lineage-specific transcriptional programs. Cdh1 inactivation resulted in shifts along the squamous differentiation trajectory associated with aberrant expression of genes central to gastrointestinal epithelial differentiation. Cytokeratin 7 (CK7), encoded by the differentiation-dependent gene Krt7, was a specific marker for early neoplastic lesions in CDH1 carriers. Our findings suggest that deregulation of developmental transcriptional programs may precede malignancy in HDGC. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Stomach Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Organoids , Single-Cell Analysis , Stomach Neoplasms/pathology , Transcriptome
4.
EMBO Rep ; 20(12): e48375, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31668005

ABSTRACT

Outcomes for metastatic Ewing sarcoma and osteosarcoma are dismal and have not changed for decades. Oxidative stress attenuates melanoma metastasis, and melanoma cells must reduce oxidative stress to metastasize. We explored this in sarcomas by screening for oxidative stress sensitizers, which identified the class I HDAC inhibitor MS-275 as enhancing vulnerability to reactive oxygen species (ROS) in sarcoma cells. Mechanistically, MS-275 inhibits YB-1 deacetylation, decreasing its binding to 5'-UTRs of NFE2L2 encoding the antioxidant factor NRF2, thereby reducing NFE2L2 translation and synthesis of NRF2 to increase cellular ROS. By global acetylomics, MS-275 promotes rapid acetylation of the YB-1 RNA-binding protein at lysine-81, blocking binding and translational activation of NFE2L2, as well as known YB-1 mRNA targets, HIF1A, and the stress granule nucleator, G3BP1. MS-275 dramatically reduces sarcoma metastasis in vivo, but an MS-275-resistant YB-1K81-to-alanine mutant restores metastatic capacity and NRF2, HIF1α, and G3BP1 synthesis in MS-275-treated mice. These studies describe a novel function for MS-275 through enhanced YB-1 acetylation, thus inhibiting YB-1 translational control of key cytoprotective factors and its pro-metastatic activity.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Bone Neoplasms/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Pyridines/therapeutic use , Sarcoma, Ewing/drug therapy , Transcription Factors/metabolism , Acetylation , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis , Oxidative Stress , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology
5.
J Pathol ; 249(3): 319-331, 2019 11.
Article in English | MEDLINE | ID: mdl-31236944

ABSTRACT

Despite being the most common childhood bone tumor, the genomic characterization of osteosarcoma remains incomplete. In particular, very few osteosarcoma metastases have been sequenced to date, critical to better understand mechanisms of progression and evolution in this tumor. We performed an integrated whole genome and exome sequencing analysis of paired primary and metastatic pediatric osteosarcoma specimens to identify recurrent genomic alterations. Sequencing of 13 osteosarcoma patients including 13 primary, 10 metastatic, and 3 locally recurring tumors revealed a highly heterogeneous mutational landscape, including cases of hypermutation and microsatellite instability positivity, but with virtually no recurrent alterations except for mutations involving the tumor suppressor genes RB1 and TP53. At the germline level, we detected alterations in multiple cancer related genes in the majority of the cohort, including those potentially disrupting DNA damage response pathways. Metastases retained only a minimal number of short variants from their corresponding primary tumors, while copy number alterations showed higher conservation. One recurrently amplified gene, KDR, was highly expressed in advanced cases and associated with poor prognosis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Osteosarcoma/genetics , Osteosarcoma/secondary , Whole Genome Sequencing , Age Factors , British Columbia , DNA Copy Number Variations , Female , Gene Amplification , Gene Dosage , Genetic Heterogeneity , Genetic Predisposition to Disease , Humans , Male , Microsatellite Instability , Mutation , Phenotype , Polymorphism, Single Nucleotide , Transcriptome , United States , Exome Sequencing
6.
J Pathol ; 244(5): 650-666, 2018 04.
Article in English | MEDLINE | ID: mdl-29293271

ABSTRACT

Altered mRNA translational control is emerging as a critical factor in cancer development and progression. Targeting specific elements of the translational machinery, such as mTORC1 or eIF4E, is emerging as a new strategy for innovative cancer therapy. While translation of most mRNAs takes place through cap-dependent mechanisms, a sub-population of cellular mRNA species, particularly stress-inducible mRNAs with highly structured 5'-UTR regions, are primarily translated through cap-independent mechanisms. Intriguingly, many of these mRNAs encode proteins that are involved in tumour cell adaptation to microenvironmental stress, and thus linked to aggressive behaviour including tumour invasion and metastasis. This necessitates a rigorous search for links between microenvironmental stress and aggressive tumour phenotypes. Under stress, cells block global protein synthesis to preserve energy while maintaining selective synthesis of proteins that support cell survival. One highly conserved mechanism to regulate protein synthesis under cell stress is to sequester mRNAs into cytosolic aggregates called stress granules (SGs), where their translation is silenced. SGs confer survival advantages and chemotherapeutic resistance to tumour cells under stress. Recently, it has been shown that genetically blocking SG formation dramatically reduces tumour invasive and metastatic capacity in vivo. Therefore, targeting SG formation might represent a potential treatment strategy to block cancer metastasis. Here, we present the critical link between selective mRNA translation, stress adaptation, SGs, and tumour progression. Further, we also explain how deciphering mechanisms of selective mRNA translation occurs under cell stress holds great promise for the identification of new targets in the treatment of cancer. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cytoplasmic Granules/genetics , Neoplasms/genetics , Protein Biosynthesis , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Stress, Physiological , Tumor Microenvironment , Animals , Cell Movement , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/pathology , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Phenotype , RNA Stability , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
7.
Clin Cancer Res ; 30(5): 1022-1037, 2024 03 01.
Article in English | MEDLINE | ID: mdl-37812652

ABSTRACT

PURPOSE: Ewing sarcoma is the second most common bone sarcoma in children, with 1 case per 1.5 million in the United States. Although the survival rate of patients diagnosed with localized disease is approximately 70%, this decreases to approximately 30% for patients with metastatic disease and only approximately 10% for treatment-refractory disease, which have not changed for decades. Therefore, new therapeutic strategies are urgently needed for metastatic and refractory Ewing sarcoma. EXPERIMENTAL DESIGN: This study analyzed 19 unique Ewing sarcoma patient- or cell line-derived xenografts (from 14 primary and 5 metastatic specimens) using proteomics to identify surface proteins for potential immunotherapeutic targeting. Plasma membranes were enriched using density gradient ultracentrifugation and compared with a reference standard of 12 immortalized non-Ewing sarcoma cell lines prepared in a similar manner. In parallel, global proteome analysis was carried out on each model to complement the surfaceome data. All models were analyzed by Tandem Mass Tags-based mass spectrometry to quantify identified proteins. RESULTS: The surfaceome and global proteome analyses identified 1,131 and 1,030 annotated surface proteins, respectively. Among surface proteins identified, both approaches identified known Ewing sarcoma-associated proteins, including IL1RAP, CD99, STEAP1, and ADGRG2, and many new cell surface targets, including ENPP1 and CDH11. Robust staining of ENPP1 was demonstrated in Ewing sarcoma tumors compared with other childhood sarcomas and normal tissues. CONCLUSIONS: Our comprehensive proteomic characterization of the Ewing sarcoma surfaceome provides a rich resource of surface-expressed proteins in Ewing sarcoma. This dataset provides the preclinical justification for exploration of targets such as ENPP1 for potential immunotherapeutic application in Ewing sarcoma. See related commentary by Bailey, p. 934.


Subject(s)
Bone Neoplasms , Sarcoma, Ewing , Sarcoma , Child , Humans , Sarcoma, Ewing/genetics , Sarcoma, Ewing/therapy , Membrane Proteins , Proteome , Proteomics , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Immunotherapy , Antigens, Neoplasm , Oxidoreductases
8.
Nat Genet ; 55(2): 255-267, 2023 02.
Article in English | MEDLINE | ID: mdl-36624343

ABSTRACT

Endometriosis is a common condition in women that causes chronic pain and infertility and is associated with an elevated risk of ovarian cancer. We profiled transcriptomes of >370,000 individual cells from endometriomas (n = 8), endometriosis (n = 28), eutopic endometrium (n = 10), unaffected ovary (n = 4) and endometriosis-free peritoneum (n = 4), generating a cellular atlas of endometrial-type epithelial cells, stromal cells and microenvironmental cell populations across tissue sites. Cellular and molecular signatures of endometrial-type epithelium and stroma differed across tissue types, suggesting a role for cellular restructuring and transcriptional reprogramming in the disease. Epithelium, stroma and proximal mesothelial cells of endometriomas showed dysregulation of pro-inflammatory pathways and upregulation of complement proteins. Somatic ARID1A mutation in epithelial cells was associated with upregulation of pro-angiogenic and pro-lymphangiogenic factors and remodeling of the endothelial cell compartment, with enrichment of lymphatic endothelial cells. Finally, signatures of ciliated epithelial cells were enriched in ovarian cancers, reinforcing epidemiologic associations between these two diseases.


Subject(s)
Endometriosis , Transcriptome , Humans , Female , Transcriptome/genetics , Endometriosis/genetics , Endometriosis/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Epithelium
9.
Pediatr Blood Cancer ; 59(7): 1215-22, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22544557

ABSTRACT

BACKGROUND: Hypoxia contributes to both physiological and pathological processes and its effects are mainly mediated through the transcription factors hypoxia-inducible factor 1α and 2α (HIF1α and HIF2α). The purpose of this study was to examine the role of these proteins in osteosarcoma progression. PROCEDURES: We developed a method to isolate primary human osteoblast cell lines. HIF1α and HIF2α expression were then compared in osteoblast and osteosarcoma cell lines under 21% oxygen (normoxia) and 1% oxygen (hypoxia). We also used hypoxia-responsive element (HRE)-driven reporter constructs in conjunction with siRNAs specific to HIF1α or HIF2α to determine the contribution of each protein to HRE-mediated transcription. Finally, we measured HIF1α expression in primary osteosarcoma tumors by immunohistochemistry. RESULTS: We found that mainly HIF1α transcript was significantly higher in osteosarcoma cell lines compared to normal osteoblasts under both normoxia and hypoxia. At the protein level, HIF1α was preferentially stabilized in osteosarcoma cell lines under both conditions. HIF1α expression was required for the observed increases in HRE activity. Finally, nuclear or nucleocytoplasmic HIF1α staining in osteosarcoma cases was associated with high-grade tumors. CONCLUSIONS: These findings point to a role for HIF1α in osteosarcoma progression and suggest that the observed differences in HIF1α oxygen dependent degradation may play an important pathophysiological role in this disease. Pediatr Blood Cancer 2012; 59: 1215-1222. © 2012 Wiley Periodicals, Inc.


Subject(s)
Bone Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Osteosarcoma/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Immunohistochemistry , Osteoblasts/metabolism , Protein Stability , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation
10.
Cell Death Differ ; 29(1): 54-64, 2022 01.
Article in English | MEDLINE | ID: mdl-34294889

ABSTRACT

Breast cancer heterogeneity has made it challenging to identify mechanisms critical to the initial stages of their genesis in vivo. Here, we sought to interrogate the role of YB-1 in newly arising human breast cancers as well as in established cell lines. In a first series of experiments, we found that short-hairpin RNA-mediated knockdown of YB-1 in MDA-MB-231 cells blocked both their local tumour-forming and lung-colonising activity in immunodeficient mice. Conversely, upregulated expression of YB-1 enhanced the poor in vivo tumorigenicity of T47D cells. We then found that YB-1 knockdown also inhibits the initial generation in mice of invasive ductal carcinomas and ductal carcinomas in situ from freshly isolated human mammary cells transduced, respectively, with KRASG12D or myristoylated-AKT1. Interestingly, increased expression of HIF1α and G3BP1, two YB-1 translational targets and elements of a stress-adaptive programme, mirrored the levels of YB-1 in both transformed primary and established MDA-MB-231 breast cancer cells.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Helicases/metabolism , Female , Humans , Mice , Poly-ADP-Ribose Binding Proteins , RNA Helicases/metabolism , RNA Recognition Motif Proteins , Transcription Factors/metabolism , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
11.
J Hazard Mater ; 403: 123575, 2021 02 05.
Article in English | MEDLINE | ID: mdl-32791477

ABSTRACT

Construction of a multipurpose yeast consortium suitable for lipid production, textile dye/effluent removal and lignin valorization is critical for both biorefinery and bioremediation. Therefore, a novel oleaginous consortium, designated as OYC-Y.BC.SH has been developed using three yeast cultures viz. Yarrowia sp. SSA1642, Barnettozyma californica SSA1518 and Sterigmatomyces halophilus SSA1511. The OYC-Y.BC.SH was able to grow on different carbon sources and accumulate lipids, with its highest lipid productivity (1.56 g/L/day) and lipase activity (170.3 U/mL) exhibited in xylose. The total saturated fatty acid content was 36.09 %, while the mono-unsaturated and poly-unsaturated fatty acids were 45.44 and 18.30 %, respectively, making OYC-Y.BC.SH valuable for biodiesel production. The OYC-Y.BC.SH showed its highest decolorization efficiency of Red HE3B dye (above 82 %) in presence of sorghum husk as agricultural co-substrate, suggesting its feasibility for simultaneous lignin valorization. The significant higher performance of OYC-Y.BC.SH on decolorizing the real dyeing effluent sample at pH 8.0 suggests its potential and suitability for degrading most of the wastewater textile effluents. Clearly, toxicological studies underline the additional advantage of using OYC-Y.BC.SH for bioremediation of industrial dyeing effluents in terms of decolorization and detoxification. A possible mechanism of Red HE3B biodegradation and ATP synthesis was also proposed.


Subject(s)
Coloring Agents , Wastewater , Basidiomycota , Biodegradation, Environmental , Biofuels , Lignin , Lipids , Saccharomycetales , Textile Industry , Textiles
12.
Oncogene ; 40(11): 1988-2001, 2021 03.
Article in English | MEDLINE | ID: mdl-33603169

ABSTRACT

Uncovering the mechanisms that underpin how tumor cells adapt to microenvironmental stress is essential to better understand cancer progression. The HACE1 (HECT domain and ankyrin repeat-containing E3 ubiquitin-protein ligase) gene is a tumor suppressor that inhibits the growth, invasive capacity, and metastasis of cancer cells. However, the direct regulatory pathways whereby HACE1 confers this tumor-suppressive effect remain to be fully elucidated. In this report, we establish a link between HACE1 and the major stress factor, hypoxia-inducible factor 1 alpha (HIF1α). We find that HACE1 blocks the accumulation of HIF1α during cellular hypoxia through decreased protein stability. This property is dependent on HACE1 E3 ligase activity and loss of Ras-related C3 botulinum toxin substrate 1 (RAC1), an established target of HACE1 mediated ubiquitinylation and degradation. In vivo, genetic deletion of Rac1 reversed the increased HIF1α expression observed in Hace1-/- mice in murine KRasG12D-driven lung tumors. An inverse relationship was observed between HACE1 and HIF1α levels in tumors compared to patient-matched normal kidney tissues, highlighting the potential pathophysiological significance of our findings. Together, our data uncover a previously unrecognized function for the HACE1 tumor suppressor in blocking HIF1α accumulation under hypoxia in a RAC1-dependent manner.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , rac1 GTP-Binding Protein/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/pathology , Mice , Mice, Knockout , Neoplasm Metastasis , Protein Stability , Signal Transduction/genetics , Tumor Hypoxia/genetics , Ubiquitination/genetics
13.
Cancer Discov ; 11(11): 2884-2903, 2021 11.
Article in English | MEDLINE | ID: mdl-34021002

ABSTRACT

Cancer cells must overcome anoikis (detachment-induced death) to successfully metastasize. Using proteomic screens, we found that distinct oncoproteins upregulate IL1 receptor accessory protein (IL1RAP) to suppress anoikis. IL1RAP is directly induced by oncogenic fusions of Ewing sarcoma, a highly metastatic childhood sarcoma. IL1RAP inactivation triggers anoikis and impedes metastatic dissemination of Ewing sarcoma cells. Mechanistically, IL1RAP binds the cell-surface system Xc - transporter to enhance exogenous cystine uptake, thereby replenishing cysteine and the glutathione antioxidant. Under cystine depletion, IL1RAP induces cystathionine gamma lyase (CTH) to activate the transsulfuration pathway for de novo cysteine synthesis. Therefore, IL1RAP maintains cyst(e)ine and glutathione pools, which are vital for redox homeostasis and anoikis resistance. IL1RAP is minimally expressed in pediatric and adult normal tissues, and human anti-IL1RAP antibodies induce potent antibody-dependent cellular cytotoxicity of Ewing sarcoma cells. Therefore, we define IL1RAP as a new cell-surface target in Ewing sarcoma, which is potentially exploitable for immunotherapy. SIGNIFICANCE: Here, we identify cell-surface protein IL1RAP as a key driver of metastasis in Ewing sarcoma, a highly aggressive childhood sarcoma. Minimal expression in pediatric and adult normal tissues nominates IL1RAP as a promising target for immunotherapy.See related commentary by Yoon and DeNicola, p. 2679.This article is highlighted in the In This Issue feature, p. 2659.


Subject(s)
Anoikis , Interleukin-1 Receptor Accessory Protein , Sarcoma, Ewing , Adult , Cell Line, Tumor , Child , Humans , Proteomics , Receptors, Interleukin-1 , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology
14.
Bioresour Technol ; 310: 123462, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32388355

ABSTRACT

In the present study, a halophilic microalgal species was isolated from a hypersaline lagoon with salinity average of 45.3‰ and identified as Dunaliella salina KSA-HS022. It was further cultivated at a salinity range of 50-250‰, applied directly to batch cultures or through stepwise increase in a semi-continuous culture. The later showed the highest biomass productivity of 0.191 g L-1 d-1 at 125‰, which represented 45.8% higher than the corresponding batch culture (control). Oxidative markers in the control cultures were significantly higher than those of the adapted culture, confirming reduction of oxidative stress by adaptation. In addition, stepwise adaptation showed the highest lipid productivity of 56.5 mg L-1 d-1 at 150‰ (39.9% higher than the corresponding control), which resulted in the highest fatty acid methyl esters productivity. Moreover, stepwise increase of salinity up to 150‰ enhanced the biodiesel characteristics, offering a new route for enhanced biodiesel production at extraordinary salinity levels.


Subject(s)
Biofuels , Microalgae , Batch Cell Culture Techniques , Biomass , Salinity
15.
Cancer Res ; 80(14): 3009-3022, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32366477

ABSTRACT

HACE1 is an E3 ubiquitin ligase with important roles in tumor biology and tissue homeostasis. Loss or mutation of HACE1 has been associated with the occurrence of a variety of neoplasms, but the underlying mechanisms have not been defined yet. Here, we report that HACE1 is frequently mutated in human lung cancer. In mice, loss of Hace1 led to enhanced progression of KRasG12D -driven lung tumors. Additional ablation of the oncogenic GTPase Rac1 partially reduced progression of Hace1-/- lung tumors. RAC2, a novel ubiquitylation target of HACE1, could compensate for the absence of its homolog RAC1 in Hace1-deficient, but not in HACE1-sufficient tumors. Accordingly, ablation of both Rac1 and Rac2 fully averted the increased progression of KRasG12D -driven lung tumors in Hace1-/- mice. In patients with lung cancer, increased expression of HACE1 correlated with reduced levels of RAC1 and RAC2 and prolonged survival, whereas elevated expression of RAC1 and RAC2 was associated with poor prognosis. This work defines HACE1 as a crucial regulator of the oncogenic activity of RAC-family GTPases in lung cancer development. SIGNIFICANCE: These findings reveal that mutation of the tumor suppressor HACE1 disrupts its role as a regulator of the oncogenic activity of RAC-family GTPases in human and murine lung cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/14/3009/F1.large.jpg.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/prevention & control , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/pathology , Cell Proliferation , Humans , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , RAC2 GTP-Binding Protein
16.
Cell Death Dis ; 10(1): 21, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622235

ABSTRACT

Osteosarcoma is a malignant bone sarcoma characterized by extensive genomic disruption and a propensity for metastatic spread. Osteoid production suggests a close relationship with normal osteoblasts, and the latter are the presumptive cell of origin of this disease. The HACE1 gene, localized to human chromosome 6q21, encodes the HACE1 HECT E3 ligase, a tumor suppressor in diverse tumors that acts in part by targeting the activated form of RAC1 GTPase for proteasomal degradation. Disruption or loss of 6q21 is relatively common in osteosarcomas, and Hace1-/-/Tp53+/- mice frequently develop osteosarcomas, in contrast to Tp53+/- mice, which do not. This suggests an unexplored link between HACE1 loss and osteosarcoma. Here we compared HACE1 expression in normal osteoblasts and osteosarcoma cell lines in vitro by western blotting and quantitative RT-PCR, and in human osteosarcoma specimens by immunohistochemistry. Both HACE1 transcript and protein levels were reduced in osteosarcoma compared to osteoblasts in vitro. Reduced HACE1 expression in osteosarcoma tumors was observed in 76% of cases and associated with high-grade lesions. Further, clonally derived pairs of high and low metastatic osteosarcoma cell lines showed significant downregulation in the high compared to corresponding low metastatic cells. Ectopic expression of HACE1 markedly inhibited anchorage-independent growth and cell motility of HACE1 osteosarcoma cell lines, and was associated with reduced RAC1 activation and decreased reactive oxygen species (ROS). Finally, HACE1 overexpression blocked osteosarcoma xenograft growth and dramatically reduced pulmonary metastases. These findings point to a potential tumor suppressor function for HACE1 in osteosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Osteosarcoma/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , HEK293 Cells , Heterografts , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Nude , Osteoblasts/metabolism , Osteosarcoma/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Transfection , rac1 GTP-Binding Protein/metabolism
17.
Commun Biol ; 2: 165, 2019.
Article in English | MEDLINE | ID: mdl-31069274

ABSTRACT

The RNA helicase EIF4A3 regulates the exon junction complex and nonsense-mediated mRNA decay functions in RNA transcript processing. However, a transcriptome-wide network definition of these functions has been lacking, in part due to the lack of suitable pharmacological inhibitors. Here we employ short-duration graded EIF4A3 inhibition using small molecule allosteric inhibitors to define the transcriptome-wide dependencies of EIF4A3. We thus define conserved cellular functions, such as cell cycle control, that are EIF4A3 dependent. We show that EIF4A3-dependent splicing reactions have a distinct genome-wide pattern of associated RNA-binding protein motifs. We also uncover an unanticipated role of EIF4A3 in the biology of RNA stress granules, which sequester and silence the translation of most mRNAs under stress conditions and are implicated in cell survival and tumour progression. We show that stress granule induction and maintenance is suppressed on the inhibition of EIF4A3, in part through EIF4A3-associated regulation of G3BP1 and TIA1 scaffold protein expression.


Subject(s)
Cell Cycle/genetics , Cytoplasmic Granules/metabolism , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/genetics , Stress, Physiological/genetics , Transcriptome , Allosteric Regulation/drug effects , Cell Cycle/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Computational Biology/methods , Cytoplasmic Granules/drug effects , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/metabolism , Gene Expression Regulation , HCT116 Cells , HeLa Cells , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Recognition Motif Proteins/genetics , RNA Recognition Motif Proteins/metabolism , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Stress, Physiological/drug effects , T-Cell Intracellular Antigen-1/genetics , T-Cell Intracellular Antigen-1/metabolism
18.
J Biosci Bioeng ; 125(2): 231-237, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29037768

ABSTRACT

Nowadays, biofuel production is a fast expanding industry and is facing a growing dilemma about a feedstock source capable of keeping up with demand. Recently, macroalgae have been attracting a wide attention as a source for biofuel. In the present study, ten macroalgae were collected and screened as biodiesel feedstocks. As a result of their high biomass production and relatively high lipid content, Ulva lactuca, Padina boryana and Ulva intestinalis showed the highest significant lipids and fatty acid methyl esters (FAMEs) areal productivities among the studied species. Saturated fatty acids (SAFs) showed insignificant differences in the selected species, with noticeably significant higher polyunsaturated fatty acids (PUFAs) content in U. lactuca by 4.2 and 3 times, with respect to P. boryana and U. intestinalis, respectively. The recorded increase in PUFAs was attributed to higher content of C16:4n-3, C18:3n-3 and C18:4n-3. By lipid fractionation, P. boryana showed significant higher concentration of neutral lipids (37.7 mg g-1 CDW, representing 46.7% of total fatty acids) in comparison to U. lactuca and U. intestinalis, which showed 16% and 17% lower neutral lipid fractions, respectively. In addition, biodiesel characteristics of the studied macroalgae complied with that of international standards. Furthermore, oil-free residual biomass can be readily converted into fermentable sugars or biogas due to its high carbohydrates content, which adds to the economics of macroalgae as biofuel feedstock. In conclusion, the present study confirmed that macroalgae represent an attractive alternative renewable feedstock for biodiesel and other biofuels.


Subject(s)
Biofuels/supply & distribution , Bioreactors , Seaweed/metabolism , Biomass , Fatty Acids/metabolism , Fermentation , Lipid Metabolism , Phaeophyceae/metabolism , Ulva/metabolism
19.
Cell Death Differ ; 24(9): 1564-1576, 2017 09.
Article in English | MEDLINE | ID: mdl-28574509

ABSTRACT

MYC family proteins are implicated in many human cancers, but their therapeutic targeting has proven challenging. MYCN amplification in childhood neuroblastoma (NB) is associated with aggressive disease and high mortality. Novel and effective therapeutic strategies are therefore urgently needed for these tumors. MYC-driven oncogenic transformation impairs cell survival under nutrient deprivation (ND), a characteristic stress condition within the tumor microenvironment. We recently identified eukaryotic Elongation Factor 2 Kinase (eEF2K) as a pivotal mediator of the adaptive response of tumor cells to ND. We therefore hypothesized that eEF2K facilitates the adaptation of MYCN amplified NB to ND, and that inhibiting this pathway can impair MYCN-driven NB progression. To test our hypothesis, we first analyzed publicly available genomic databases and tissue microarrays for eEF2K expression in NB, and for links between eEF2K, MYCN, and clinical outcome in NB. Effects of eEF2K inhibition were evaluated on survival of MYCN amplified versus non-amplified NB cell lines under ND. Finally, NB xenograft mouse models were used to confirm in vitro observations. Our results indicate that high eEF2K expression and activity are strongly predictive of poor outcome in NB, and correlates significantly with MYCN amplification. Inhibition of eEF2K markedly decreases survival of MYCN amplified NB cell lines in vitro under ND. Growth of MYCN amplified NB xenografts is markedly impaired by eEF2K knockdown, particularly under caloric restriction. In summary, eEF2K protects MYCN overexpressing NB cells from ND in vitro and in vivo, highlighting this kinase as a critical mediator of the adaptive response of MYCN amplified NB cells to metabolic stress.


Subject(s)
Elongation Factor 2 Kinase/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , RNA, Messenger/metabolism , Animals , Blotting, Western , Cell Line , Elongation Factor 2 Kinase/genetics , Female , Flow Cytometry , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, SCID , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Tissue Array Analysis
20.
Article in English | MEDLINE | ID: mdl-27656243

ABSTRACT

Research on marine natural products as potential anticancer agents is still limited. In the present study, an aqueous extract of a Canadian marine microalgal preparation was assessed for anticancer activities using various assays and cell lines of human cancers, including lung, prostate, stomach, breast, and pancreatic cancers, as well as an osteosarcoma. In vitro, the microalgal extract exhibited marked anticolony forming activity. In addition, it was more toxic, as indicated by increased apoptosis, to nonadherent cells (grown in suspension) than to adherent cells. In vivo, an antimetastatic effect of the extract was observed in NOD-SCID mice carrying subrenal capsule xenografts of PC3 prostate cancer cells. The results of the present study suggest that the antimetastatic effect of the aqueous microalgal extract is based on inhibition of colony forming ability of cancer cells and the preferential killing of suspended cancer cells. Further research aimed at identification of the molecular basis of the anticancer activities of the microalgal extract appears to be warranted.

SELECTION OF CITATIONS
SEARCH DETAIL