Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Int J Surg Pathol ; 32(3): 551-555, 2024 May.
Article in English | MEDLINE | ID: mdl-37394760

ABSTRACT

TFEB-amplified renal cell carcinoma (RCC), which belongs to the MITF family of RCC, is characterized by genomic amplification at the 6p21.1 locus where the TFEB gene is located. The vascular endothelial growth factor A and cyclin D3 genes are also located at this same locus. When tumors lack classic morphologic features, they may be classified as "RCC not otherwise specified (NOS)." However, it is increasingly important to accurately diagnose the RCC subtype to define the patient's individual prognosis and select the subsequent therapeutic modalities, which now include targeted agents. Therefore, knowledge of the diagnostic features of TFEB-altered RCCs, such as t(6;11) RCCs and TFEB-amplified RCCs, is critical for identifying these tumors. Herein, we present an interesting case of TFEB-amplified RCC that was initially diagnosed as RCC NOS on biopsy of a renal tumor in a community practice setting with available molecular findings demonstrating CCND3 amplification. The genetic abnormality was "accidentally" detected due to the amplification of the colocated CCND3 gene at the 6p21 locus of the TFEB gene on a limited genetic sequencing panel. This case highlights the importance of molecular tests in accurately diagnosing RCC and carefully interpreting molecular findings in the context of histomorphologic features.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Gene Amplification , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Translocation, Genetic , Biomarkers, Tumor/genetics , Cyclin D3/genetics , Cyclin D3/metabolism
2.
Clin Transl Oncol ; 26(3): 698-708, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37540409

ABSTRACT

PURPOSE: There is compelling evidence that long-stranded non-coding RNAs (lncRNAs) play an important role in the progression of hepatocellular carcinoma (HCC). The aim of this study was to investigate the role of lncRNA XXYLT1 antisense-2 (XXYLT1-AS2) in HCC progression. METHODS: Real-time PCR was used to assess the levels of XXYLT1-AS2 in plasma from HCC and normal patients. Cell proliferation, apoptosis, migration, and invasion were monitored, and tumor xenografts were established to investigate the biological functions of XXYLT1-AS2 by gain-of-function and loss-of-function studies in vitro and in vivo, the expression of autophagy biomarkers and transcriptional factor EB (TFEB) was examined by immunoprecipitation, ubiquitination assays, and western blotting. Autophagy inhibitor, 3-methyladenine (3MA), and proteasome inhibitor, MG132, were used to verify the role of autophagy in HCC progression and the effect of XXYLT1-AS2 on TFEB ubiquitination, respectively. RESULTS: In this study, we identified that lncRNA XXYLT1-AS2 is highly expressed in HCC plasma and promotes tumor growth in vivo. In functional studies, it was found that silent expression of XXYLT1-AS2 inhibited HCC proliferation, migration, invasion, and activated autophagy of HCC cells, which were attenuated by autophagy inhibitor, 3MA. Mechanistically, XXYLT1-AS2 decreased the protein level of TFEB through promoting its degradation by ubiquitin proteasome pathway. CONCLUSION: XXYLT1-AS2 plays an oncogenic role in HCC progression through inhibition of autophagy via promoting the degradation of TFEB, and thus could be a novel target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Carcinoma, Hepatocellular/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Liver Neoplasms/pathology , Cell Line, Tumor , Autophagy/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , MicroRNAs/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
3.
J Cell Physiol ; 238(5): 954-965, 2023 05.
Article in English | MEDLINE | ID: mdl-37013375

ABSTRACT

The mechanisms of autophagy have been related to Alzheimer's disease (AD) pathogenesis by the endosomal-lysosomal system, having a critical function in forming amyloid-ß (Aß) plaques. Nevertheless, the exact mechanisms mediating disease pathogenesis remain unclear. The transcription factor EB (TFEB), a primary transcriptional autophagy regulator, improves gene expression, mediating lysosome function, autophagic flux, and autophagosome biogenesis. In this review, we present for the first time the hypothesis of how TFEB, autophagy, and mitochondrial function are interconnected in AD, providing a logical foundation for unraveling the critical role of chronic physical exercise in this process. Aerobic exercise training promotes Adiponectin Receptor 1 (AdipoR1)/AMP-activated protein kinase (AMPK)/TFEB axis activation in the brain of the AD animal model, which contributes to alleviated Aß deposition and neuronal apoptosis while improving cognitive function. Moreover, TFEB upregulates Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and nuclear factor erythroid 2-related factor 2 (NRF-2), improving mitochondrial biogenesis and redox status. In addition, tissue contraction activates calcineurin in skeletal muscle, which induces TFEB nuclear translocation, raising the hypothesis that the same would occur in the brain. Thus, a deep and comprehensive exploration of the TFEB could provide new directions and strategies for preventing AD. We conclude that chronic exercise can be an effective TFEB activator, inducing autophagy and mitochondrial biogenesis, representing a potential nonpharmacological strategy contributing to brain health.


Subject(s)
Alzheimer Disease , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Animals , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Exercise , Lysosomes/metabolism , Muscle, Skeletal/metabolism
4.
Clin Transl Oncol ; 24(7): 1333-1346, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35118587

ABSTRACT

PURPOSE: Translocation renal cell carcinoma (tRCC) is a subtype that occurs predominantly in children and young individuals. Metastatic tRCC occurring in young patients is more aggressive than that occurring in older patients, and there are still no effective therapies. Organoids can mimic original tissues and be assessed by high-throughput screening (HTS). We aimed to utilize patient-derived organoids and HTS to screen drugs that can be repurposed for metastatic tRCC with PRCC-TFE3 fusion. METHODS: Tumor tissues were obtained from treatment-naïve metastatic tRCC patients who underwent surgery. Histopathology and fluorescence in situ hybridization (FISH) confirmed the tRCC. Organoids derived from the dissected tissues were cultured and verified by FISH and RNA-seq. HTS was performed to seek promising drugs, and potential mechanisms were explored by RNA-seq and cell-based studies. RESULTS: We successfully established a metastatic tRCC organoid with PRCC-TFE3 fusion, a common fusion subtype, and its characteristics were verified by histopathology, FISH, and RNA-seq. An HTS assay was developed, and the robustness was confirmed. A compound library of 1816 drugs was screened. Eventually, axitinib, crizotinib, and JQ-1 were selected for further validation and were found to induce cell cycle arrest and apoptosis. RNA-seq analyses of posttreatment organoids indicated that crizotinib induced significant changes in autophagy-related genes, consistent with the potential pathogenesis of tRCC. CONCLUSIONS: We established and validated organoids derived from tissues dissected from a patient with metastatic tRCC with PRCC-TFE3 fusion and achieved the HTS process for the first time. Crizotinib might be a targeted therapy worthy of exploration in the clinic for metastatic tRCC with PRCC-TFE3 fusion. Such organoid and HTS assays may represent a promising model system in translational research assisting in the development of clinical strategies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Aged , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Crizotinib/pharmacology , Humans , In Situ Hybridization, Fluorescence , Kidney Neoplasms/diagnosis , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Organoids , Translocation, Genetic
5.
Int J Mol Sci ; 22(8)2021 Apr 19.
Article in English | MEDLINE | ID: mdl-33921734

ABSTRACT

Niemann-Pick type C disease (NPCD) is a lysosomal storage disease (LSD) characterized by abnormal cholesterol accumulation in lysosomes, impaired autophagy flux, and lysosomal dysfunction. The activation of transcription factor EB (TFEB), a master lysosomal function regulator, reduces the accumulation of lysosomal substrates in LSDs where the degradative capacity of the cells is compromised. Genistein can pass the blood-brain barrier and activate TFEB. Hence, we investigated the effect of TFEB activation by genistein toward correcting the NPC phenotype. We show that genistein promotes TFEB translocation to the nucleus in HeLa TFEB-GFP, Huh7, and SHSY-5Y cells treated with U18666A and NPC1 patient fibroblasts. Genistein treatment improved lysosomal protein expression and autophagic flux, decreasing p62 levels and increasing those of the LC3-II in NPC1 patient fibroblasts. Genistein induced an increase in ß-hexosaminidase activity in the culture media of NPC1 patient fibroblasts, suggesting an increase in lysosomal exocytosis, which correlated with a decrease in cholesterol accumulation after filipin staining, including cells treated with U18666A and NPC1 patient fibroblasts. These results support that genistein-mediated TFEB activation corrects pathological phenotypes in NPC models and substantiates the need for further studies on this isoflavonoid as a potential therapeutic agent to treat NPCD and other LSDs with neurological compromise.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Genistein/therapeutic use , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/metabolism , Androstenes/therapeutic use , Animals , Blotting, Western , Cell Line, Tumor , Cholesterol/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Humans , Lysosomal Storage Diseases , Lysosomes/metabolism , Niemann-Pick C1 Protein/metabolism
6.
Cells ; 9(5)2020 05 14.
Article in English | MEDLINE | ID: mdl-32423042

ABSTRACT

The adverse environmental conditions found in the periodontium during periodontitis pathogenesis stimulate local autophagy responses, mainly due to a continuous inflammatory response against the dysbiotic subgingival microbiome. The junctional epithelium represents the main site of the initial interaction between the host and the dysbiotic biofilm. Here, we investigated the role of autophagy in junctional epithelium keratinocytes (JEKs) in response to Aggregatibacter actinomycetemcomitans or its purified lipopolysaccharides (LPS). Immunofluorescence confocal analysis revealed an extensive nuclear translocation of transcription factor EB (TFEB) and consequently, an increase in autophagy markers and LC3-turnover assessed by immunoblotting and qRT-PCR. Correspondingly, challenged JEKs showed a punctuate cytosolic profile of LC3 protein contrasting with the diffuse distribution observed in untreated controls. Three-dimensional reconstructions of confocal images displayed a close association between intracellular bacteria and LC3-positive vesicles. Similarly, a close association between autophagic vesicles and the protein p62 was observed in challenged JEKs, indicating that p62 is the main adapter protein recruited during A. actinomycetemcomitans infection. Finally, the pharmacological inhibition of autophagy significantly increased the number of bacteria-infected cells as well as their death, similar to treatment with LPS. Our results indicate that A. actinomycetemcomitans infection induces autophagy in JEKs, and this homeostatic process has a cytoprotective effect on the host cells during the early stages of infection.


Subject(s)
Aggregatibacter actinomycetemcomitans/physiology , Autophagy , Epithelial Attachment/pathology , Keratinocytes/microbiology , Keratinocytes/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Biomarkers/metabolism , Cell Count , Cell Line , Cell Nucleus/metabolism , Cell Survival , Humans , Imaging, Three-Dimensional , Lipopolysaccharides/isolation & purification , Models, Biological , Protein Transport , Sequestosome-1 Protein/metabolism
7.
FASEB J ; 34(1): 1602-1619, 2020 01.
Article in English | MEDLINE | ID: mdl-31914620

ABSTRACT

Endurance exercise begun with reduced muscle glycogen stores seems to potentiate skeletal muscle protein abundance and gene expression. However, it is unknown whether this greater signaling responses is due to performing two exercise sessions in close proximity-as a first exercise session is necessary to reduce the muscle glycogen stores. In the present study, we manipulated the recovery duration between a first muscle glycogen-depleting exercise and a second exercise session, such that the second exercise session started with reduced muscle glycogen in both approaches but was performed either 2 or 15 hours after the first exercise session (so-called "twice-a-day" and "once-daily" approaches, respectively). We found that exercise twice-a-day increased the nuclear abundance of transcription factor EB (TFEB) and nuclear factor of activated T cells (NFAT) and potentiated the transcription of peroxisome proliferator-activated receptor-É£ coactivator 1-alpha (PGC-1α), peroxisome proliferator-activated receptor-alpha (PPARα), and peroxisome proliferator-activated receptor beta/delta (PPARß/δ) genes, in comparison with the once-daily exercise. These results suggest that part of the elevated molecular signaling reported with previous "train-low" approaches might be attributed to performing two exercise sessions in close proximity. The twice-a-day approach might be an effective strategy to induce adaptations related to mitochondrial biogenesis and fat oxidation.


Subject(s)
Biomarkers/metabolism , Exercise/physiology , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/physiology , AMP-Activated Protein Kinases/metabolism , Adaptation, Physiological/physiology , Adult , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Cell Nucleus/physiology , Cross-Over Studies , Glycogen/metabolism , Humans , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , NFATC Transcription Factors/metabolism , Organelle Biogenesis , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
8.
J Mol Med (Berl) ; 97(10): 1491-1506, 2019 10.
Article in English | MEDLINE | ID: mdl-31401659

ABSTRACT

Mesenchymal stromal cells (MSCs) are frequently recruited to tumor sites to play a part in the tumor microenvironment (TME). However, their real impact on cancer cell behavior remains obscure. Here we investigated the effects of human adipose-derived stromal cell (hADSC) secretome in autophagy of glioblastoma (GBM), as a way to better comprehend how hADSCs influence the TME. GBM U-87 MG cells were treated with conditioned medium (CM) from hADSCs and autophagic flux was evaluated. hADSC CM treatment blocked the autophagic flux in tumor cells, as indicated by the accumulation of autophagosomes in the cytosol, the high LC3-II and p62/SQSTM1 protein levels, and the lack of increase in the amount of acidic vesicular organelles. These effects were further detected in other GBM cell lines tested and also in co-cultures of hADSCs and U-87 MG. hADSC CM did not compromise lysosomal acidification; however, it was able to activate mTORC1 signaling and, as a consequence, led to a decrease in the nuclear translocation of TFEB, a master transcriptional regulator of lysosomal biogenesis and autophagy, thereby contributing to a defective autophagic process. hADSCs secrete transforming growth factor beta 1 (TGFß1) and this cytokine is an important mediator of CM effects on autophagy. A comprehensive knowledge of MSC roles in tumor biology is of great importance to shed light on the complex dialog between these cells and to explore such interactions therapeutically. The present results help to elucidate the paracrine effects of MSCs in tumors and bring attention to the potential to be explored in MSC secretome. KEY MESSAGES: hADSC secretome specifically affects the biology of GBM cells. hADSCs block the late steps of autophagic flux in GBM cells. hADSC secretome activates mTORC1 signaling and reduces TFEB nuclear translocation in GBM cells.


Subject(s)
Autophagy/drug effects , Culture Media, Conditioned/pharmacology , Stromal Cells/metabolism , Tumor Microenvironment/drug effects , Active Transport, Cell Nucleus/drug effects , Adipose Tissue/cytology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects
9.
Scand J Clin Lab Invest ; 79(3): 218-224, 2019 May.
Article in English | MEDLINE | ID: mdl-30813849

ABSTRACT

Most quantitative real-time PCR (qPCR) detection methods use two types of chemistries to measure the expression levels of ChREBP isoforms, hydrolysis probes for ChREBPα and SYBR Green for ChREBPß. Hydrolysis probes are not available to determine the ChREBPß isoform. The aim of this study was to develop a qPCR assay based only on hydrolysis probes for both ChREBP isoforms. Liver and adipose tissue biopsies from patients undergoing elective cholecystectomy surgery were used to perform qPCR. To validate this assay, the results were compared with sequencing and High Resolution Melting (HRM) PCR assays. Direct sequencing was used to determine the sequence showing site where ChREBPß presents its specific splicing (1 b exon/2 exon) in order to design the primers and the probe. We developed a qPCR assay to determine the ChREBP isoforms expression based on hydrolysis probes. It assays showed good efficiency (95.50%, on average), high reproducibility, and a strong linear correlation (R2 ≥ 0.99) for tissues tested. HRM analysis confirmed the specificity of the primers and the result of this assay matched (100%) with the outcomes obtained by sequencing and qPCR. Also, we obtained the ChREBPß sequence showing exon 1b spliced to exon 2, bypassing exon 1a, and retaining the remainder of the ChREBPα exons. Based on the use of hydrolysis probes, our method can efficiently identify the expression of both ChREBP isoforms. Thus, the comparability of the qPCR results using a single chemistry (hydrolysis probes) to discriminate between both ChREBP isoforms was possible.


Subject(s)
Adipose Tissue/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Liver/metabolism , Real-Time Polymerase Chain Reaction/methods , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Humans , Omentum/metabolism , Protein Isoforms/metabolism , Subcutaneous Tissue/metabolism
10.
Nucleic Acids Res ; 45(W1): W344-W349, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28407117

ABSTRACT

Peptides are extensively used to characterize functional or (linear) structural aspects of receptor-ligand interactions in biological systems, e.g. SH2, SH3, PDZ peptide-recognition domains, the MHC membrane receptors and enzymes such as kinases and phosphatases. NNAlign is a method for the identification of such linear motifs in biological sequences. The algorithm aligns the amino acid or nucleotide sequences provided as training set, and generates a model of the sequence motif detected in the data. The webserver allows setting up cross-validation experiments to estimate the performance of the model, as well as evaluations on independent data. Many features of the training sequences can be encoded as input, and the network architecture is highly customizable. The results returned by the server include a graphical representation of the motif identified by the method, performance values and a downloadable model that can be applied to scan protein sequences for occurrence of the motif. While its performance for the characterization of peptide-MHC interactions is widely documented, we extended NNAlign to be applicable to other receptor-ligand systems as well. Version 2.0 supports alignments with insertions and deletions, encoding of receptor pseudo-sequences, and custom alphabets for the training sequences. The server is available at http://www.cbs.dtu.dk/services/NNAlign-2.0.


Subject(s)
Algorithms , Neural Networks, Computer , Peptides/chemistry , Software , Amino Acid Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Databases, Protein , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , HLA-A1 Antigen/chemistry , HLA-A1 Antigen/metabolism , HLA-B7 Antigen/chemistry , HLA-B7 Antigen/metabolism , HLA-B8 Antigen/chemistry , HLA-B8 Antigen/metabolism , HLA-DRB1 Chains/chemistry , HLA-DRB1 Chains/metabolism , Humans , Internet , Ligands , Peptides/metabolism , Protein Binding , Sequence Alignment , Trans-Activators/chemistry , Trans-Activators/metabolism
11.
Protoplasma ; 254(3): 1399-1410, 2017 May.
Article in English | MEDLINE | ID: mdl-27696021

ABSTRACT

Plant growth-promoting rhizobacteria are natural inhabitants of roots, colonize diverse monocot and dicot species, and affect several functional traits such as root architecture, adaptation to adverse environments, and protect plants from pathogens. N,N-dimethyl-hexadecylamine (C16-DMA) is a rhizobacterial amino lipid that modulates the postembryonic development of several plants, likely as part of volatile blends. In this work, we evaluated the bioactivity of C16-DMA and other related N,N-dimethyl-amines with varied length and found that inhibition of primary root growth was related to the length of the acyl chain. C16-DMA inhibited primary root growth affecting cell division and elongation, while promoting lateral root formation and root hair growth and density in Arabidopsis thaliana (Arabidopsis) wild-type (WT) seedlings. Interestingly, C16-DMA induced the expression of the jasmonic acid (JA)-responsive gene marker pLOX2:uidA, while JA-related mutants jar1, coi1-1, and myc2 affected on JA biosynthesis and perception, respectively, are compromised in C16-DMA responses. Comparison of auxin-regulated gene expression, root architectural changes in WT, and auxin-related mutants aux1-7, tir1/afb2/afb3, and arf7-1/arf19-1 to C16-DMA shows that the C16-DMA effects occur independently of auxin signaling. Together, these results reveal a novel class of aminolipids modulating root organogenesis via crosstalk with the JA signaling pathway.


Subject(s)
Arabidopsis/metabolism , Cyclopentanes/metabolism , Methylamines/pharmacology , Morphogenesis/drug effects , Oxylipins/metabolism , Plant Roots/growth & development , Arabidopsis/growth & development , Arabidopsis Proteins/biosynthesis , Arabidopsis Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Division/drug effects , Indoleacetic Acids/metabolism , Lipoxygenases/biosynthesis , Methylamines/chemistry , Methylamines/metabolism , Nucleotidyltransferases/metabolism , Plant Roots/cytology , Plant Roots/microbiology , Seedlings/metabolism , Signal Transduction
12.
Cell Tissue Res ; 364(3): 647-660, 2016 06.
Article in English | MEDLINE | ID: mdl-26742767

ABSTRACT

Fibrosis occurs in skeletal muscle under various pathophysiological conditions such as Duchenne muscular dystrophy (DMD), a devastating disease characterized by fiber degeneration that results in progressive loss of muscle mass, weakness and increased extracellular matrix (ECM) accumulation. Fibrosis is also observed after skeletal muscle denervation and repeated cycles of damage followed by regeneration. The ECM is synthesized largely by fibroblasts in the muscle connective tissue under normal conditions. Myofibroblasts, cells that express α-smooth muscle actin (α-SMA), play a role in many tissues affected by fibrosis. In skeletal muscle, fibro/adipogenic progenitors (FAPs) that express cell-surface platelet-derived growth factor receptor-α (PDGFR-α) and the transcription factor Tcf4 seem to be responsible for connective tissue synthesis and are good candidates for the origin of myofibroblasts. We show that cells positive for Tcf4 and PDGFR-α are expressed in skeletal muscle under normal conditions and are increased in various skeletal muscles of mdx mice, a murine model for DMD, wild type muscle after sciatic denervation and muscle subjected to chronic damage. These cells co-label with the myofibroblast marker α-SMA in dystrophic muscle but not in normal tissue. The Tcf4-positive cells lie near macrophages mainly concentrated in dystrophic necrotic-regenerating foci. The close proximity of Tcf4-positive cells to inflammatory cells and their previously described role in muscle regeneration might reflect an active interaction between these cell types and growth factors, possibly resulting in a muscular regenerative or fibrotic condition.


Subject(s)
Adipogenesis , Biomarkers/metabolism , Cell Differentiation , Connective Tissue/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myofibroblasts/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Count , Chronic Disease , Denervation , Fibrosis , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Muscle, Skeletal/innervation , Muscular Dystrophy, Animal/pathology , Necrosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Transcription Factor 4 , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Oxid Med Cell Longev ; 2015: 482582, 2015.
Article in English | MEDLINE | ID: mdl-26583058

ABSTRACT

Mitochondria are essential organelles for eukaryotic homeostasis. Although these organelles possess their own DNA, the vast majority (>99%) of mitochondrial proteins are encoded in the nucleus. This situation makes systems that allow the communication between mitochondria and the nucleus a requirement not only to coordinate mitochondrial protein synthesis during biogenesis but also to communicate eventual mitochondrial malfunctions, triggering compensatory responses in the nucleus. Mitochondria-to-nucleus retrograde signaling has been described in various organisms, albeit with differences in effector pathways, molecules, and outcomes, as discussed in this review.


Subject(s)
Mitochondria/metabolism , Signal Transduction , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mitochondrial Proteins/metabolism , Multiprotein Complexes/metabolism , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
Genet Mol Res ; 14(4): 14009-18, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26535715

ABSTRACT

The Wnt pathway has been implicated in the initiation, progression, and metastasis of lung cancer. T cell factor 4, a member of TCF/LEF family, acts as a transcriptional factor for Wnt pathways in lung cancer. Increasing amounts of evidence have shown that TCF-4 has multiple alternative splicing isoforms with transactivation or transrepression activity toward the Wnt pathway. Here, we found the presence of multiple TCF-4 isoforms in lung cancer cell lines and in normal bronchial epithelial cells. TCF-4K isoform expression was significantly decreased in lung cancer cells compared with normal bronchial epithelial cells and was identified as a transcriptional suppressor of the Wnt pathway in non-small cell lung carcinoma (NSCLC). Overexpression of TCF-4K significantly inhibited the proliferation and migration of NSCLC cells. Collectively, our data indicate that TCF-4K functions as a tumor suppressor in NSCLC by down-regulating the Wnt pathway.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Bronchi/cytology , Bronchi/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cloning, Molecular , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Lung Neoplasms/genetics , Neoplasm Metastasis , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor 4 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Wnt Signaling Pathway/genetics
15.
Genet Mol Res ; 14(1): 1220-8, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25730060

ABSTRACT

Free-range production system is increasingly being used in poultry breeding and feed production in many countries. The objective of the current experiment was to evaluate the effects of different raising systems on fat-related traits and mRNA levels of liver lipogenesis genes in Erlang Mountainous chicken. Each of 10 birds (91 day old) from caged, indoor-floor housed, and free-range housing systems was slaughtered, and fat-related traits, live body weight (BW), subcutaneous fat thickness (SFT), abdominal fat weight (AFW), abdominal fat percentage (AFP), and intramuscular fat content were determined. The mRNA levels of liver X receptor α, carbohydrate response element-binding protein (ChREBP), sterol regulatory element-binding protein-1 (SREBP1), and fatty acid synthase were detected. The caged chicken exhibited significantly higher BW, SFT, and AFW than those of free-ranged chicken (P < 0.05). All the 4 genes had a similar expression pattern, and they showed the highest level in caged chicken, while the lowest level was found in free-ranged chicken. Association analysis indicated that there were significant (P < 0.05) or highly significant (P < 0.01) positive correlations between the mRNA levels of ChREBP, SREBP1, and fat traits of SFT, AFW, and AFP. Thus, we deduced that increased fat deposition in caged chicken was probably induced by increased gene expression of ChREBP and SREBP1 in the liver.


Subject(s)
Abdominal Fat/metabolism , Avian Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Chickens/genetics , Housing, Animal , Sterol Regulatory Element Binding Protein 1/metabolism , Subcutaneous Fat/metabolism , Animals , Avian Proteins/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Body Weight , Fatty Acid Synthases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Liver/metabolism , Male , Real-Time Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics
16.
Free Radic Biol Med ; 81: 30-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25578655

ABSTRACT

Mitochondrial retrograde signaling is a communication pathway between the mitochondrion and the nucleus that regulates the expression of a subset of nuclear genes that codify mitochondrial proteins, mediating cell response to mitochondrial dysfunction. In Saccharomyces cerevisiae, the pathway depends on Rtg1p and Rtg3p, which together form the transcription factor that regulates gene expression, and Rtg2p, an activator of the pathway. Here, we provide novel studies aimed at assessing the functional impact of the lack of RTG-dependent signaling on mitochondrial activity. We show that mutants defective in RTG-dependent retrograde signaling present higher oxygen consumption and reduced hydrogen peroxide release in the stationary phase compared to wild-type cells. Interestingly, RTG mutants are less able to decompose hydrogen peroxide or maintain viability when challenged with hydrogen peroxide. Overall, our results indicate that RTG signaling is involved in the hormetic induction of antioxidant defenses and stress resistance.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction , Active Transport, Cell Nucleus/drug effects , Adaptation, Physiological/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Nucleus/drug effects , Cell Nucleus/genetics , Gene Expression Regulation, Fungal , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/drug effects , Mitochondria/genetics , Oxidative Stress , Phosphorylation , Protein Transport/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
17.
Mol Cell Biol ; 35(4): 737-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25512609

ABSTRACT

Lipid droplets (LDs) are intracellular structures that regulate neutral lipid homeostasis. In mammals, LD synthesis is inhibited by rapamycin, a known inhibitor of the mTORC1 pathway. In Saccharomyces cerevisiae, LD dynamics are modulated by the growth phase; however, the regulatory pathways involved are unknown. Therefore, we decided to study the role of the TORC1 pathway on LD metabolism in S. cerevisiae. Interestingly, rapamycin treatment resulted in a fast LD replenishment and growth inhibition. The discovery that osmotic stress (1 M sorbitol) also induced LD synthesis but not growth inhibition suggested that the induction of LDs in yeast is not a secondary response to reduced growth. The induction of LDs by rapamycin was due to increased triacylglycerol but not sterol ester synthesis. Induction was dependent on the TOR downstream effectors, the PP2A-related phosphatase Sit4p and the regulatory protein Tap42p. The TORC1-controlled transcriptional activators Gln3p, Gat1p, Rtg1p, and Rtg3p, but not Msn2p and Msn4p, were required for full induction of LDs by rapamycin. Furthermore, we show that the deletion of Gln3p and Gat1p transcription factors, which are activated in response to nitrogen availability, led to abnormal LD dynamics. These results reveal that the TORC1 pathway is involved in neutral lipid homeostasis in yeast.


Subject(s)
Gene Expression Regulation, Fungal , Lipid Droplets/metabolism , Phosphatidylinositol 3-Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cholesterol Esters/metabolism , GATA Transcription Factors/deficiency , GATA Transcription Factors/genetics , Lipid Droplets/chemistry , Lipid Droplets/drug effects , Lipid Metabolism/drug effects , Osmotic Pressure , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Sirolimus/pharmacology , Sorbitol/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency , Transcription Factors/metabolism , Triglycerides/biosynthesis
18.
J Interferon Cytokine Res ; 34(11): 858-69, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24950290

ABSTRACT

Interferon-α2b (IFN-α2b) reduces proliferation and increases apoptosis in hepatocellular carcinoma cells by decreasing ß-catenin/TCF4/Smads interaction. Forkhead box O-class 3a (FoxO3a) participates in proliferation and apoptosis and interacts with ß-catenin and Smads. FoxO3a is inhibited by Akt, IκB kinase ß (IKKß), and extracellular-signal-regulated kinase (Erk), which promote FoxO3a sequestration in the cytosol, and accumulates in the nucleus upon phosphorylation by c-Jun N-terminal kinase (JNK) and p38 mitogen-activated kinase (p38 MAPK). We analyzed FoxO3a subcellular localization, the participating kinases, FoxO3a/ß-catenin/Smads association, and FoxO3a target gene expression in IFN-α2b-stimulated HepG2/C3A and Huh7 cells. Total FoxO3a and Akt-phosphorylated FoxO3a levels decreased in the cytosol, whereas total FoxO3a levels increased in the nucleus upon IFN-α2b stimulus. IFN-α2b reduced Akt, IKKß, and Erk activation, and increased JNK and p38 MAPK activation. p38 MAPK inhibition blocked IFN-α2b-induced FoxO3a nuclear localization. IFN-α2b enhanced FoxO3a association with ß-catenin and Smad2/3/7. Two-step coimmunoprecipitation experiments suggest that these proteins coexist in the same complex. The expression of several FoxO3a target genes increased with IFN-α2b. FoxO3a knockdown prevented the induction of these genes, suggesting that FoxO3a acts as mediator of IFN-α2b action. Results suggest a ß-catenin/Smads switch from TCF4 to FoxO3a. Such events would contribute to the IFN-α2b-mediated effects on cellular proliferation and apoptosis. These results demonstrate new mechanisms for IFN-α action, showing the importance of its application in antitumorigenic therapies.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cell Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Immunotherapy/methods , Interferon-alpha/pharmacology , Smad Proteins/metabolism , beta Catenin/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Hepatocellular/immunology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hep G2 Cells , Humans , Protein Binding/drug effects , Protein Transport/drug effects , RNA, Small Interfering/genetics , Transcription Factor 4 , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
PLoS One ; 8(12): e82457, 2013.
Article in English | MEDLINE | ID: mdl-24349289

ABSTRACT

Neuroblastoma is the most common extracranial tumor and a major cause of infant cancer mortality worldwide. Despite its importance, little is known about its molecular mechanisms. A striking feature of this tumor is its clinical heterogeneity. Possible outcomes range from aggressive invasion to other tissues, causing patient death, to spontaneous disease regression or differentiation into benign ganglioneuromas. Several efforts have been made in order to find tumor progression markers. In this work, we have reconstructed the neuroblastoma regulatory network using an information-theoretic approach in order to find genes involved in tumor progression and that could be used as outcome predictors or as therapeutic targets. We have queried the reconstructed neuroblastoma regulatory network using an aggressive neuroblastoma metastasis gene signature in order to find its master regulators (MRs). MRs expression profiles were then investigated in other neuroblastoma datasets so as to detect possible clinical significance. Our analysis pointed MAX as one of the MRs of neuroblastoma progression. We have found that higher MAX expression correlated with favorable patient outcomes. We have also found that MAX expression and protein levels were increased during neuroblastoma SH-SY5Y cells differentiation. We propose that MAX is involved in neuroblastoma progression, possibly increasing cell differentiation by means of regulating the availability of MYC:MAX heterodimers. This mechanism is consistent with the results found in our SH-SY5Y differentiation protocol, suggesting that MAX has a more central role in these cells differentiation than previously reported. Overexpression of MAX has been identified as anti-tumorigenic in other works, but, to our knowledge, this is the first time that the link between the expression of this gene and malignancy was verified under physiological conditions.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Neuroblastoma/genetics , Neuroblastoma/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cluster Analysis , Disease Progression , Gene Expression Profiling , Humans , Models, Biological , Neuroblastoma/metabolism , Neuroblastoma/mortality , Signal Transduction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL