Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Nat Commun ; 15(1): 4097, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755144

ABSTRACT

Angiogenesis, the growth of new blood vessels from pre-existing vasculature, is essential for the development of new organ systems, but transcriptional control of angiogenesis remains incompletely understood. Here we show that FOXC1 is essential for retinal angiogenesis. Endothelial cell (EC)-specific loss of Foxc1 impairs retinal vascular growth and expression of Slc3a2 and Slc7a5, which encode the heterodimeric CD98 (LAT1/4F2hc) amino acid transporter and regulate the intracellular transport of essential amino acids and activation of the mammalian target of rapamycin (mTOR). EC-Foxc1 deficiency diminishes mTOR activity, while administration of the mTOR agonist MHY-1485 rescues perturbed retinal angiogenesis. EC-Foxc1 expression is required for retinal revascularization and resolution of neovascular tufts in a model of oxygen-induced retinopathy. Foxc1 is also indispensable for pericytes, a critical component of the blood-retina barrier during retinal angiogenesis. Our findings establish FOXC1 as a crucial regulator of retinal vessels and identify therapeutic targets for treating retinal vascular disease.


Subject(s)
Blood-Retinal Barrier , Endothelial Cells , Forkhead Transcription Factors , Retinal Neovascularization , Animals , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Mice , Endothelial Cells/metabolism , Blood-Retinal Barrier/metabolism , TOR Serine-Threonine Kinases/metabolism , Pericytes/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Retinal Vessels/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Mice, Knockout , Mice, Inbred C57BL , Retina/metabolism , Male , Angiogenesis
2.
Thorac Cancer ; 15(17): 1369-1384, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38720472

ABSTRACT

BACKGROUND: Solute carrier family 34 member 2 (SLC34A2) has been implicated in the development of various malignancies. However, the clinical significance and underlying molecular mechanisms of SLC34A2 in esophageal squamous cell carcinoma (ESCC) remain elusive. METHODS: Western blotting, quantitative real-time PCR and immunohistochemistry were utilized to evaluate the expression levels of SLC34A2 mRNA/protein in ESCC cell lines or tissues. Kaplan-Meier curves were employed for survival analysis. CCK-8, colony formation, EdU and xenograft tumor model assays were conducted to determine the impact of SLC34A2 on ESCC cell proliferation. Cell cycle was examined using flow cytometry. RNA-sequencing and enrichment analysis were carried out to explore the potential signaling pathways. The autophagic flux was evaluated by western blotting, mRFP-GFP-LC3 reporter system and transmission electron microscopy. Immunoprecipitation and mass spectrometry were utilized for identification of potential SLC34A2-interacting proteins. Cycloheximide (CHX) chase and ubiquitination assays were conducted to test the protein stability. RESULTS: The expression of SLC34A2 was significantly upregulated in ESCC and correlated with unfavorable clinicopathologic characteristics particularly the Ki-67 labeling index and poor prognosis of ESCC patients. Overexpression of SLC34A2 promoted ESCC cell proliferation, while silencing SLC34A2 had the opposite effect. Moreover, SLC34A2 induced autophagy to promote ESCC cell proliferation, whereas inhibition of autophagy suppressed the proliferation of ESCC cells. Further studies showed that SLC34A2 interacted with an autophagy-related protein STX17 to promote autophagy and proliferation of ESCC cells by inhibiting the ubiquitination and degradation of STX17. CONCLUSIONS: These findings indicate that SLC34A2 may serve as a prognostic biomarker for ESCC.


Subject(s)
Autophagy , Cell Proliferation , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Mice , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/genetics , Animals , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Female , Male , Prognosis , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Mice, Nude , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Xenograft Model Antitumor Assays
3.
J Biol Chem ; 299(12): 105409, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37918802

ABSTRACT

Maintenance of the proteasome requires oxidative phosphorylation (ATP) and mitigation of oxidative damage, in an increasingly dysfunctional relationship with aging. SLC3A2 plays a role on both sides of this dichotomy as an adaptor to SLC7A5, a transporter of branched-chain amino acids (BCAA: Leu, Ile, Val), and to SLC7A11, a cystine importer supplying cysteine to the synthesis of the antioxidant glutathione. Endurance in mammalian muscle depends in part on oxidation of BCAA; however, elevated serum levels are associated with insulin resistance and shortened lifespans. Intriguingly, the evolution of modern birds (Neoaves) has entailed the purging of genes including SLC3A2, SLC7A5, -7, -8, -10, and SLC1A4, -5, largely removing BCAA exchangers and their interacting Na+/Gln symporters in pursuit of improved energetics. Additional gene purging included mitochondrial BCAA aminotransferase (BCAT2), pointing to reduced oxidation of BCAA and increased hepatic conversion to triglycerides and glucose. Fat deposits are anhydrous and highly reduced, maximizing the fuel/weight ratio for prolonged flight, but fat accumulation in muscle cells of aging humans contributes to inflammation and senescence. Duplications of the bidirectional α-ketoacid transporters SLC16A3, SLC16A7, the cystine transporters SLC7A9, SLC7A11, and N-glycan branching enzymes MGAT4B, MGAT4C in Neoaves suggests a shift to the transport of deaminated essential amino acid, and stronger mitigation of oxidative stress supported by the galectin lattice. We suggest that Alfred Lotka's theory of natural selection as a maximum power organizer (PNAS 8:151,1922) made an unusually large contribution to Neoave evolution. Further molecular analysis of Neoaves may reveal novel rewiring with applications for human health and longevity.


Subject(s)
Birds , Evolution, Molecular , Longevity , Animals , Humans , Amino Acids, Branched-Chain/metabolism , Cystine/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Liver/metabolism , Longevity/genetics , Birds/genetics , Birds/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Selection, Genetic
4.
J Exp Clin Cancer Res ; 42(1): 200, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37559159

ABSTRACT

BACKGROUND: Advanced colorectal cancer (CRC) is difficult to treat. For that reason, the development of novel therapeutics is necessary. Here we describe a potentially actionable plasma membrane target, the amino acid transporter protein subunit CD98hc. METHODS: Western blot and immunohistochemical analyses of CD98hc protein expression were carried out on paired normal and tumoral tissues from patients with CRC. Immunofluorescence and western studies were used to characterize the action of a DM1-based CD98hc-directed antibody-drug conjugate (ADC). MTT and Annexin V studies were performed to evaluate the effect of the anti-CD98hc-ADC on cell proliferation and apoptosis. CRISPR/Cas9 and shRNA were used to explore the specificity of the ADC. In vitro analyses of the antitumoral activity of the anti-CD98hc-ADC on 3D patient-derived normal as well as tumoral organoids were also carried out. Xenografted CRC cells and a PDX were used to analyze the antitumoral properties of the anti-CD98hc-ADC. RESULTS: Genomic as well proteomic analyses of paired normal and tumoral samples showed that CD98hc expression was significantly higher in tumoral tissues as compared to levels of CD98hc present in the normal colonic tissue. In human CRC cell lines, an ADC that recognized the CD98hc ectodomain, reached the lysosomes and exerted potent antitumoral activity. The specificity of the CD98hc-directed ADC was demonstrated using CRC cells in which CD98hc was decreased by shRNA or deleted using CRISPR/Cas9. Studies in patient-derived organoids verified the antitumoral action of the anti-CD98hc-ADC, which largely spared normal tissue-derived colon organoids. In vivo studies using xenografted CRC cells or patient-derived xenografts confirmed the antitumoral activity of the anti-CD98hc-ADC. CONCLUSIONS: The studies herewith reported indicate that CD98hc may represent a novel ADC target that, upon well-designed clinical trials, could be used to increase the therapeutic armamentarium against CRC.


Subject(s)
Colorectal Neoplasms , Fusion Regulatory Protein 1, Heavy Chain , Humans , Fusion Regulatory Protein 1, Heavy Chain/genetics , Proteomics , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , RNA, Small Interfering , Cell Line, Tumor
5.
Int J Med Sci ; 20(8): 1060-1078, 2023.
Article in English | MEDLINE | ID: mdl-37484811

ABSTRACT

Background: Solute Carrier Family 3 Member 2 (SLC3A2) is a member of the solute carrier family that plays pivotal roles in regulation of intracellular calcium levels and transports L-type amino acids. However, there are insufficient scientific researches on the prognostic and immunological roles of SLC3A2 in breast cancer (BC) and whether everolimus regulates novel SLC3A2 related molecular mechanism in the immuno-oncology context of the tumor microenvironment (TME), therefore, we see a necessity to conduct the current in silico and biological experimental study. Methods: Using diverse online databases, we investigated the role of SLC3A2 in therapy response, clinicopathological characteristics, tumor immune infiltration, genetic alteration, methylation and single cell sequencing in BC. WB, Co-IP, cell proliferation assay, Edu staining, ROS and GSH assay and in vivo tumor xenograft assays were performed to verify FKBP1A/SLC3A2 axis in everolimus inducing ferroptosis of breast cancer. Co-cultures and IL-9 ELISA were performed to demonstrate the T lymphocyte function. Results: We demonstrated that SLC3A2 was aberrantly expressed among various BC cohorts. Our results also suggested that SLC3A2 expression was associated with chemotherapeutic outcome in BC patients. Our results further indicated that SLC3A2 was associated with tumor infiltration of cytotoxic T cell but not other immune cells among BC TME. The alterations in SLC3A2 gene had a significant correlation to relapse free survival and contributed a significant impact on BC tumor mutational burden. Finally, SLC3A2 was illustrated to be expressed in diverse BC cellular populations at single cell level, and negatively linked to angiogenesis, inflammation and quiescence, but positively correlated with other functional phenotypes. Noteworthily, everolimus (a targeted therapy drug for BC) related protein, FK506-binding protein 1A (FKBP1A) was found to bind with SLC3A2, and negatively regulated SLC3A2 expression during the processes of everolimus inducing ferroptosis of BC cells and promoting anti-proliferation of Th9 lymphocytes. Conclusions: Altogether, our study strongly implies that SLC3A2 is an immuno-oncogenic factor and FKBP1A/SLC3A2 axis would provide insights for a novel immunotherapy approach for the treatment of BC in the context of TME.


Subject(s)
Breast Neoplasms , Ferroptosis , Humans , Female , Everolimus/pharmacology , Everolimus/therapeutic use , Tacrolimus Binding Protein 1A/metabolism , Ferroptosis/genetics , Neoplasm Recurrence, Local , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Tumor Microenvironment/genetics , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
6.
Cell Mol Biol Lett ; 27(1): 40, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35596131

ABSTRACT

BACKGROUND: In patients with acute aortic dissection (AAD), increased vascular smooth muscle cell (VSMC) apoptosis has been found. Human cytomegalovirus (HCMV)-miR-US33-5p was significantly increased in the plasma of patients with AAD. However, the roles of miR-US33-5p in human aortic VSMC (HA-VSMC) apoptosis remain to be elucidated. METHODS: In the current study, cell apoptosis was analyzed by flow cytometry, cell proliferation by CCK-8 assay, and differentially expressed genes by RNA sequencing. Luciferase reporter assay was used for binding analysis between miR-US33-5p and endothelial PAS domain protein 1 (EPAS1), and EPAS1 and amino acid transporter heavy chain, member 2 (SLC3A2). The enrichment degree of SLC3A2 promoter DNA was analyzed by chromatin immunoprecipitation assay. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and immunoblotting were performed for measuring messenger RNA (mRNA) and protein levels, respectively. RESULTS: It was found that HCMV infection inhibited proliferation but promoted HA-VSMC apoptosis by upregulating HCMV-miR-US33-5p. Transfection of HCMV-miR-US33-5p mimics the significant effect on several signaling pathways including integrin signaling as shown in the RNA sequencing data. Western blotting analysis confirmed that HCMV-miR-US33-5p mimics suppression of the activity of key factors of the integrin signal pathway including FAK, AKT, CAS, and Rac. Mechanistic study showed that HCMV-miR-US33-5p bound to the 3'-untranslated region of EPAS1 to suppress its expression, leading to suppression of SLC3A2 expression, which ultimately promoted cell apoptosis and inhibited cell proliferation. This was confirmed by the findings that silencing EPAS1 significantly reduced the SLC3A2 expression and inhibited proliferation and key factors of integrin signal pathway. CONCLUSIONS: HCMV-miR-US33-5p suppressed proliferation, key factors of integrin signal pathway, and EPAS1/SLC3A2 expression, but promoted HA-VSMC apoptosis. These findings highlighted the importance of HCMV-miR-US33-5p/EPAS1/SCL3A2 signaling and may provide new insights into therapeutic strategies for AAD.


Subject(s)
Aortic Dissection , Cytomegalovirus , MicroRNAs , Myocytes, Smooth Muscle , Aortic Dissection/metabolism , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation/genetics , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Humans , Integrins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism
7.
Commun Biol ; 5(1): 403, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35501367

ABSTRACT

Estrogen receptor (ER) positive breast cancer represents 75% of all breast cancers in women. Although patients with ER+ cancers receive endocrine therapies, more than 30% develop resistance and succumb to the disease, highlighting the need to understand endocrine resistance. Here we show an unexpected role for the cell polarity protein SCRIB as a tumor-promoter and a regulator of endocrine resistance in ER-positive breast cancer cells. SCRIB expression is induced by estrogen signaling in a MYC-dependent manner. SCRIB interacts with SLC3A2, a heteromeric component of leucine amino acid transporter SLC7A5. SLC3A2 binds to the N-terminus of SCRIB to facilitate the formation of SCRIB/SLC3A2/LLGL2/SLC7A5 quaternary complex required for membrane localization of the amino acid transporter complex. Both SCRIB and SLC3A2 are required for cell proliferation and tamoxifen resistance in ER+ cells identifying a new role for the SCRIB/SLC3A2 complex in ER+ breast cancer.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Fusion Regulatory Protein 1, Heavy Chain , Membrane Proteins , Tamoxifen , Tumor Suppressor Proteins , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Proliferation , Cytoskeletal Proteins , Estrogens , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Humans , Large Neutral Amino Acid-Transporter 1 , Membrane Proteins/genetics , Receptors, Estrogen , Tamoxifen/pharmacology , Tumor Suppressor Proteins/genetics
8.
Hereditas ; 159(1): 6, 2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35057861

ABSTRACT

OBJECTIVE: This study aimed to explore the mRNA and protein expression of SLC3A2 in laryngeal carcinoma cells and tissues, and functional regulatory mechanism of SLC3A2 in cell ferroptosis of laryngeal carcinoma. METHODS: We chose the key gene-SLC3A2 of DEGs from TCGA by bioinformatics analysis, and then we constructed stable knockdown of SLC3A2 in laryngeal carcinoma cells. MTT assay and clonogenic assay were used to determine cell viability and cell growth, respectively. The mRNA and protein expression were determined by RT-qPCR and western blotting, respectively. Xenograft tumor model was used to determine the role of SLC3A2 in tumor growth. RESULTS: The results of limma analysis recovered that 92 genes were involved in both upregulated DEGs and high risk of poor prognosis, whereas 36 genes were involved in both downregulated DEGs and low risk of poor prognosis. Pathway enrichment analysis indicated that mTOR signaling pathway and ferroptosis exerted a role in regulating these intersection genes. Moreover, SLC3A2 is a key gene in ferroptosis in laryngeal carcinoma. SLC3A2 is highly expressed in laryngeal carcinoma tissues and cells. Patients with high SLC3A2 expression exerted poor survival. SLC3A2 deficiency inhibited cell proliferation and foci formation. Furthermore, knockdown of SLC3A2 expression induced the efficacy of ferroptosis and suppressed ferroptosis related proteins expression. Mechanically, SLC3A2 deficiency facilitated ferroptosis through upregulating the expression of mTOR and P70S6K, whereas inhibited p-mTOR and p-P70S6K expression in laryngeal carcinoma cells. SLC3A2 deficiency inhibited tumorigenesis in nude mice. CONCLUSION: Our study suggests that SLC3A2 negatively regulates ferroptosis through mTOR pathway in laryngeal carcinoma.


Subject(s)
Carcinoma , Ferroptosis , Fusion Regulatory Protein 1, Heavy Chain/genetics , Laryngeal Neoplasms/pathology , Animals , Cell Proliferation , Humans , Laryngeal Neoplasms/genetics , Mice , Mice, Nude , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
9.
Pathol Res Pract ; 229: 153730, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34942513

ABSTRACT

L-type amino acid transporter (LAT1) is a neutral amino acid transporter, forming a heterodimer complex with the CD98 heavy chain (CD98hc). In this study, we studied the expression profiles of LAT1 and CD98hc in colorectal cancer (CRC) and its precursor lesions. Transcription levels of CD98hc and LAT1 were significantly increased in CRC compared to the matched normal mucosa. CD98hc and LAT1 expression showed no significant correlations with cancer stem cell markers and intestinal stem cell markers, whereas both had positive correlations with Wnt target genes, AXIN2, and EPHB2, suggesting an association with aberrant Wnt signaling activation. Immunohistochemical analysis revealed that CD98hc and LAT1 are not expressed in normal colonic mucosa and various benign lesions including hyperplastic polyps and sessile and traditional serrated adenomas. CD98hc and LAT1 expressions began to appear in tubular adenomas and further increased in carcinomas. Of interest, CD98hc expression decreased during lymph node metastasis. Survival analysis demonstrated that CD98hc and LAT1 have no significant prognostic effect in CRCs. In conclusion, CD98hc and LAT1 are not normally expressed in colonic mucosa and most benign lesions. Their expression began to appear in tubular adenomas and further increased during the adenoma-to-carcinoma transition. CD98hc expression decreased while metastasizing to regional lymph nodes. However, CD98hc and LAT1 expressions had no prognostic value in patients with CRC.


Subject(s)
Colorectal Neoplasms/genetics , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation, Neoplastic , Large Neutral Amino Acid-Transporter 1/genetics , Female , Humans , Male , Middle Aged , Prognosis , Survival Analysis
10.
Molecules ; 26(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34684812

ABSTRACT

It is known that 4F2hc and rBAT are the heavy subunits of the heteromeric amino acid transporters (HATs). These heavy subunits are N-glycosylated proteins, with an N-terminal domain, one transmembrane domain and a bulky extracellular domain (ectodomain) that belongs to the α-amylase family. The heavy subunits are covalently linked to a light subunit from the SLC7 family, which is responsible for the amino acid transport activity, forming a heterodimer. The functions of 4F2hc and rBAT are related mainly to the stability and trafficking of the HATs in the plasma membrane of vertebrates, where they exert the transport activity. Moreover, 4F2hc is a modulator of integrin signaling, has a role in cell fusion and it is overexpressed in some types of cancers. On the other hand, some mutations in rBAT are found to cause the malfunctioning of the b0,+ transport system, leading to cystinuria. The ectodomains of 4F2hc and rBAT share both sequence and structure homology with α-amylase family members. Very recently, cryo-EM has revealed the structure of several HATs, including the ectodomains of rBAT and 4F2hc. Here, we analyze available data on the ectodomains of rBAT and 4Fhc and their relationship with the α-amylase family. The physiological relevance of this relationship remains largely unknown.


Subject(s)
Amino Acid Transport Systems/chemistry , alpha-Glucosidases/chemistry , Amino Acid Sequence , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems, Basic/chemistry , Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Neutral/chemistry , Amino Acid Transport Systems, Neutral/genetics , Animals , Catalytic Domain , Cryoelectron Microscopy , Fusion Regulatory Protein 1, Heavy Chain/chemistry , Fusion Regulatory Protein 1, Heavy Chain/genetics , Humans , Models, Molecular , Protein Domains , Protein Multimerization , Protein Subunits , alpha-Glucosidases/genetics
11.
Nat Microbiol ; 6(8): 991-999, 2021 08.
Article in English | MEDLINE | ID: mdl-34294905

ABSTRACT

More than one-third of the world's population is exposed to Plasmodium vivax malaria, mainly in Asia1. P. vivax preferentially invades reticulocytes (immature red blood cells)2-4. Previous work has identified 11 parasite proteins involved in reticulocyte invasion, including erythrocyte binding protein 2 (ref. 5) and the reticulocyte-binding proteins (PvRBPs)6-10. PvRBP2b binds to the transferrin receptor CD71 (ref. 11), which is selectively expressed on immature reticulocytes12. Here, we identified CD98 heavy chain (CD98), a heteromeric amino acid transporter from the SLC3 family (also known as SLCA2), as a reticulocyte-specific receptor for the PvRBP2a parasite ligand using mass spectrometry, flow cytometry, biochemical and parasite invasion assays. We characterized the expression level of CD98 at the surface of immature reticulocytes (CD71+) and identified an interaction between CD98 and PvRBP2a expressed at the merozoite surface. Our results identify CD98 as an additional host membrane protein, besides CD71, that is directly associated with P. vivax reticulocyte tropism. These findings highlight the potential of using PvRBP2a as a vaccine target against P. vivax malaria.


Subject(s)
Erythrocytes/parasitology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Malaria, Vivax/metabolism , Plasmodium vivax/metabolism , Antigens, CD , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Erythrocytes/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Host-Parasite Interactions , Humans , Malaria, Vivax/blood , Malaria, Vivax/genetics , Plasmodium vivax/genetics , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Transferrin , Reticulocytes/metabolism , Reticulocytes/parasitology
12.
Sci Rep ; 11(1): 11478, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075107

ABSTRACT

The 4F2 cell-surface antigen heavy chain (4F2hc) forms a heterodimeric complex with L-type amino acid transporter 1 (LAT1) and transports large neutral essential amino acids. However, in contrast to the traditional role of LAT1 in various cancers, the role of 4F2hc has largely remained unknown. The role of 4F2hc in prostate cancer was studied. Treatment of C4-2 cells with si4F2hc was found to suppress cellular growth, migratory and invasive abilities, with this effect occurring through the cell cycle, with a significant decrease in S phase and a significant increase in G0/G1 phase, suggesting cell cycle arrest. In addition, it was proven by RNA seq that the key to 4F2hc's impact on cancer is SKP2. si4F2hc upregulates the protein expression of cyclin-dependent kinase inhibitors (P21cip1, P27kip1) through the downstream target SKP2. Furthermore, the expression of 4F2hc and LAT1 in prostate cancer cells suggests the importance of 4F2hc. Multivariate analysis showed that high 4F2hc expression was an independent prognostic factor for progression-free survival (HR 11.54, p = 0.0357). High 4F2hc was related to the clinical tumour stage (p = 0.0255) and Gleason score (p = 0.0035). Collectively, 4F2hc contributed significantly to prostate cancer (PC) progression. 4F2hc may be a novel marker and therapeutic target in PC.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/metabolism , G1 Phase , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Resting Phase, Cell Cycle , S-Phase Kinase-Associated Proteins/metabolism , Cell Line, Tumor , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , S-Phase Kinase-Associated Proteins/genetics
13.
Sci Rep ; 11(1): 9092, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907288

ABSTRACT

Neonatal encephalopathy due to hypoxia-ischemia is associated with adverse neurodevelopmental effects. The involvement of branched chain amino acids (BCAAs) in this is largely unexplored. Transport of BCAAs at the plasma membrane is facilitated by SLC7A5/SLC3A2, which increase with hypoxia. We hypothesized that hypoxia would alter BCAA transport and metabolism in the neonatal brain. We investigated this using an organotypic forebrain slice culture model with, the SLC7A5/SLC3A2 inhibitor, 2-Amino-2-norbornanecarboxylic acid (BCH) under normoxic or hypoxic conditions. We subsequently analysed the metabolome and candidate gene expression. Hypoxia was associated with increased expression of SLC7A5 and SLC3A2 and an increased tissue abundance of BCAAs. Incubation of slices with 13C-leucine confirmed that this was due to increased cellular uptake. BCH had little effect on metabolite abundance under normoxic or hypoxic conditions. This suggests hypoxia drives increased cellular uptake of BCAAs in the neonatal mouse forebrain, and membrane mediated transport through SLC7A5 and SLC3A2 is not essential for this process. This indicates mechanisms exist to generate the compounds required to maintain essential metabolism in the absence of external nutrient supply. Moreover, excess BCAAs have been associated with developmental delay, providing an unexplored mechanism of hypoxia mediated pathogenesis in the developing forebrain.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/metabolism , Hypoxia/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Prosencephalon/physiology , Adaptation, Biological , Amino Acids, Branched-Chain/metabolism , Animals , Animals, Newborn , Biological Transport , Carboxylic Acids/pharmacology , Cell Hypoxia , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation , Hypoxia/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Male , Mice, Inbred C57BL , Norbornanes/pharmacology , Organ Culture Techniques , Prosencephalon/drug effects
14.
Genet Test Mol Biomarkers ; 24(12): 812-818, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33290143

ABSTRACT

Background: Previous evidence has suggested that norepinephrine transporter (NET) gene (solute carrier family 6, member 2 [SLC6A2]) polymorphisms are involved in antidepressant response. Specifically, the polymorphism T-182C (rs2242446) located in the promoter region of SLC6A2 has been found to be associated with antidepressant response in multiple ethnic backgrounds. However, the results are inconsistent. Moreover, few studies have focused on how this T-182C polymorphism might regulate SLC6A2 promoter function. Methods: In this study, luciferase reporter assays were performed to examine the functional significance of the T-182C polymorphism. In addition, we performed a meta-analysis to explore whether this genetic variant is significantly involved in the antidepressant response. Results: We found that the -182(T) allele significantly increased promoter function compared to the C allele based on luciferase reporter assays. For the meta-analysis, six articles were identified that explored the relationship between the NET T-182C polymorphisms and antidepressant response. This study revealed no significant association between these polymorphisms and response to antidepressants (OR = 1.23, 95% CI = 0.77 - 1.97, p = 0.38 for T-182C). Conclusions: The T-182C polymorphism enhances promoter activity, but may not be associated with antidepressant response.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder, Major/genetics , Fusion Regulatory Protein 1, Heavy Chain/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Asian People/genetics , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/ethnology , Drug Resistance/genetics , Genes, Reporter , Genetic Predisposition to Disease , Genetic Vectors/genetics , HEK293 Cells , Humans , Norepinephrine/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Protein Sci ; 29(12): 2398-2407, 2020 12.
Article in English | MEDLINE | ID: mdl-33016372

ABSTRACT

System xc - is an amino acid antiporter that imports L-cystine into cells and exports intracellular L-glutamate, at a 1:1 ratio. As L-cystine is an essential precursor for glutathione synthesis, system xc - supports tumor cell growth through glutathione-based oxidative stress resistance and is considered as a potential therapeutic target for cancer treatment. System xc - consists of two subunits, the light chain subunit SLC7A11 (xCT) and the heavy chain subunit SLC3A2 (also known as CD98hc or 4F2hc), which are linked by a conserved disulfide bridge. Although the recent structures of another SLC7 member, L-type amino acid transporter 1 (LAT1) in complex with CD98hc, have provided the structural basis toward understanding the amino acid transport mechanism, the detailed molecular mechanism of xCT remains unknown. To revealthe molecular mechanism, we performed single-particle analyses of the xCT-CD98hc complex. As wild-type xCT-CD98hc displayed poor stability and could not be purified to homogeneity, we applied a consensus mutagenesis approach to xCT. The consensus mutated construct exhibited increased stability as compared to the wild-type, and enabled the cryoelectron microscopy (cryo-EM) map to be obtained at 6.2 Å resolution by single-particle analysis. The cryo-EM map revealed sufficient electron density to assign secondary structures. In the xCT structure, the hash and arm domains are well resolved, whereas the bundle domain shows some flexibility. CD98hc is positioned next to the xCT transmembrane domain. This study provides the structural basis of xCT, and our consensus-based strategy could represent a good choice toward solving unstable protein structures.


Subject(s)
Amino Acid Transport System y+/chemistry , Amino Acid Transport System y+/ultrastructure , Cryoelectron Microscopy , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Fusion Regulatory Protein 1, Heavy Chain/chemistry , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Heavy Chain/ultrastructure , HEK293 Cells , Humans , Mutagenesis , Protein Domains , Protein Stability , Protein Structure, Secondary , Sf9 Cells , Spodoptera
16.
J Immunol ; 205(10): 2883-2892, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33077644

ABSTRACT

CD98, which is required for the rapid proliferation of both normal and cancer cells, and MET, the hepatocyte growth factor receptor, are potential targets for therapeutic antitumor Abs. In this study, we report that the antiproliferative activity of a prototype anti-CD98 Ab, UM7F8, is due to Ab-induced membrane-associated ring CH (MARCH) E3 ubiquitin ligase-mediated ubiquitination and downregulation of cell surface CD98. MARCH1-mediated ubiquitination of CD98 is required for UM7F8's capacity to reduce CD98 surface expression and its capacity to inhibit the proliferation of murine T cells. Similarly, CD98 ubiquitination is required for UM7F8's capacity to block the colony-forming ability of murine leukemia-initiating cells. To test the potential generality of the paradigm that MARCH E3 ligases can mediate the antiproliferative response to antitumor Abs, we examined the potential effects of MARCH proteins on responses to emibetuzumab, an anti-MET Ab currently in clinical trials for various cancers. We report that MET surface expression is reduced by MARCH1, 4, or 8-mediated ubiquitination and that emibetuzumab-induced MET ubiquitination contributes to its capacity to downregulate MET and inhibit human tumor cell proliferation. Thus, MARCH E3 ligases can act as cofactors for antitumor Abs that target cell surface proteins, suggesting that the MARCH protein repertoire of cells is a determinant of their response to such Abs.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/pharmacology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Neoplasms/drug therapy , Ubiquitin-Protein Ligases/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/therapeutic use , Cell Proliferation/drug effects , Fusion Regulatory Protein 1, Heavy Chain/antagonists & inhibitors , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/immunology , Gene Knockout Techniques , HeLa Cells , Humans , Jurkat Cells , Mice , Mice, Knockout , Neoplasms/immunology , Neoplasms/pathology , Proteolysis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/immunology
17.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32601108

ABSTRACT

Chlamydia trachomatis infection of the human fallopian tubes can lead to damaging inflammation and scarring, ultimately resulting in infertility. To study the human cellular responses to chlamydial infection, researchers have frequently used transformed cell lines that can have limited translational relevance. We developed a primary human fallopian tube epithelial cell model based on a method previously established for culture of primary human bronchial epithelial cells. After protease digestion and physical dissociation of excised fallopian tubes, epithelial cell precursors were expanded in growth factor-containing medium. Expanded cells were cryopreserved to generate a biobank of cells from multiple donors and cultured at an air-liquid interface. Culture conditions stimulated cellular differentiation into polarized mucin-secreting and multiciliated cells, recapitulating the architecture of human fallopian tube epithelium. The polarized and differentiated cells were infected with a clinical isolate of C. trachomatis, and inclusions containing chlamydial developmental forms were visualized by fluorescence and electron microscopy. Apical secretions from infected cells contained increased amounts of proteins associated with chlamydial growth and replication, including transferrin receptor protein 1, the amino acid transporters SLC3A2 and SLC1A5, and the T-cell chemoattractants CXCL10, CXCL11, and RANTES. Flow cytometry revealed that chlamydial infection induced cell surface expression of T-cell homing and activation proteins, including ICAM-1, VCAM-1, HLA class I and II, and interferon gamma receptor. This human fallopian tube epithelial cell culture model is an important tool with translational potential for studying cellular responses to Chlamydia and other sexually transmitted pathogens.


Subject(s)
Epithelial Cells/immunology , Gene Expression Regulation/immunology , Host Microbial Interactions/immunology , T-Lymphocytes/immunology , Adult , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Biomarkers/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Chlamydia Infections/genetics , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/immunology , Epithelial Cells/microbiology , Fallopian Tubes/cytology , Fallopian Tubes/surgery , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Host Microbial Interactions/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Models, Biological , Primary Cell Culture , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Receptors, Transferrin/genetics , Receptors, Transferrin/immunology , Salpingectomy , T-Lymphocytes/microbiology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , Interferon gamma Receptor
18.
Commun Biol ; 3(1): 130, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188932

ABSTRACT

Comprehensive development is critical for gut macrophages being essential for the intestinal immune system. However, the underlying mechanisms of macrophage development in the colon remain elusive. To investigate the function of branched-chain amino acids in the development of gut macrophages, an inducible knock-out mouse model for the branched-chain amino acid transporter CD98hc in CX3CR1+ macrophages was generated. The relatively selective deletion of CD98hc in macrophage populations leads to attenuated severity of chemically-induced colitis that we assessed by clinical, endoscopic, and histological scoring. Single-cell RNA sequencing of colonic lamina propria macrophages revealed that conditional deletion of CD98hc alters the "monocyte waterfall"-development to MHC II+ macrophages. The change in the macrophage development after deletion of CD98hc is associated with increased apoptotic gene expression. Our results show that CD98hc deletion changes the development of colonic macrophages.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Colitis/metabolism , Colon/metabolism , Fusion Regulatory Protein 1, Heavy Chain/deficiency , Intestinal Mucosa/metabolism , Macrophages/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colon/ultrastructure , Crohn Disease/diagnosis , Crohn Disease/genetics , Crohn Disease/metabolism , Dextran Sulfate , Disease Models, Animal , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation , Humans , Intestinal Mucosa/ultrastructure , Macrophages/ultrastructure , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phenotype , RNA-Seq , Single-Cell Analysis , Young Adult
19.
Theranostics ; 10(5): 2172-2187, 2020.
Article in English | MEDLINE | ID: mdl-32089738

ABSTRACT

Enhanced amino acid supply and dysregulated integrin signaling constitute two hallmarks of cancer and are pivotal for metastatic transformation of cells. In line with its function at the crossroads of both processes, overexpression of CD98hc is clinically observed in various cancer malignancies, thus rendering it a promising tumor target. Methods: We describe the development of Anticalin proteins based on the lipocalin 2 (Lcn2) scaffold against the human CD98hc ectodomain (hCD98hcED) using directed evolution and protein design. X-ray structural analysis was performed to identify the epitope recognized by the lead Anticalin candidate. The Anticalin - with a tuned plasma half-life using PASylation® technology - was labeled with 89Zr and investigated by positron emission tomography (PET) of CD98-positive tumor xenograft mice. Results: The Anticalin P3D11 binds CD98hc with picomolar affinity and recognizes a protruding loop structure surrounded by several glycosylation sites within the solvent exposed membrane-distal part of the hCD98hcED. In vitro studies revealed specific binding activity of the Anticalin towards various CD98hc-expressing human tumor cell lines, suggesting broader applicability in cancer research. PET/CT imaging of mice bearing human prostate carcinoma xenografts using the optimized and 89Zr-labeled Anticalin demonstrated strong and specific tracer accumulation (8.6 ± 1.1 %ID/g) as well as a favorable tumor-to-blood ratio of 11.8. Conclusion: Our findings provide a first proof of concept to exploit CD98hc for non-invasive biomedical imaging. The novel Anticalin-based αhCD98hc radiopharmaceutical constitutes a promising tool for preclinical and, potentially, clinical applications in oncology.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/genetics , Heterografts/diagnostic imaging , Integrins/genetics , Lipocalin-2/metabolism , Protein Engineering/methods , Animals , Carcinoma , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Male , Mice, SCID , Positron Emission Tomography Computed Tomography/methods , Precision Medicine , Prostatic Neoplasms/pathology , Radiopharmaceuticals , Signal Transduction
20.
Int J Mol Sci ; 21(4)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32093034

ABSTRACT

The majority of breast cancers are oestrogen-receptor-positive (ER+) and are subject to endocrine therapy; however, an unpredictable subgroup of patients will develop resistance to endocrine therapy. The SLC7A5/SLC3A2 complex is a major route for the transport of large neutral essential amino acids through the plasma membrane. Alterations in the expression and function of those amino-acid transporters lead to metabolic reprogramming, which contributes to the tumorigenesis and drug resistance. This study aims to assess the effects and roles of SLC7A5/SLC3A2 co-expression in predicting responses to endocrine therapy in patients with ER+ breast cancer. The biological and clinical impact of SLC7A5/SLC3A2 co-expression was assessed in large annotated cohorts of ER+/HER2- breast cancer with long-term follow-up at the mRNA and protein levels. In vitro experiments were conducted to investigate the effect of SLC7A5/SLC3A2 knockdown in the proliferation of cancer cells and to the sensitivity to tamoxifen. We found that proliferation-related genes are highly expressed in a subgroup of patients with high SLC7A5/SLC3A2, and knockdown of SLC7A5/SLC3A2 decreased proliferation of ER+ breast cancer cells. In patients treated with endocrine therapy, high SLC7A5/SLC3A2 co-expression was associated with poor patient outcome, and depletion of SLC7A5/SLC3A2 using siRNA increased the sensitivity of breast cancer cells to tamoxifen. On the basis of our findings, SLC7A5/SLC3A2 co-expression has the potential of identifying a subgroup of ER+/HER2- breast cancer patients who fail to benefit from endocrine therapy and could guide the choice of other alternative therapies.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Large Neutral Amino Acid-Transporter 1/metabolism , Receptors, Estrogen/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cohort Studies , Drug Resistance, Neoplasm/genetics , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Large Neutral Amino Acid-Transporter 1/genetics , Middle Aged , Neoplasm Metastasis/genetics , Prognosis , RNA, Small Interfering , Regression Analysis , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...