Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Cell Rep ; 43(3): 113912, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38446659

ABSTRACT

In this study, we explore the dynamic process of colorectal cancer progression, emphasizing the evolution toward a more metastatic phenotype. The term "evolution" as used in this study specifically denotes the phenotypic transition toward a higher metastatic potency from well-formed glandular structures to collective invasion, ultimately resulting in the development of cancer cell buddings at the invasive front. Our findings highlight the spatial correlation of this evolution with tumor cell senescence, revealing distinct types of senescent tumor cells (types I and II) that play different roles in the overall cancer progression. Type I senescent tumor cells (p16INK4A+/CXCL12+/LAMC2-/MMP7-) are identified in the collective invasion region, whereas type II senescent tumor cells (p16INK4A+/CXCL12+/LAMC2+/MMP7+), representing the final evolved form, are prominently located in the partial-EMT region. Importantly, type II senescent tumor cells associate with local invasion and lymph node metastasis in colorectal cancer, potentially affecting patient prognosis.


Subject(s)
Colorectal Neoplasms , Matrix Metalloproteinase 7 , Humans , Matrix Metalloproteinase 7/genetics , Cellular Senescence/genetics , Phenotype , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology
2.
Nat Commun ; 14(1): 7619, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37993434

ABSTRACT

The biological process of aging is thought to result in part from accumulation of senescent cells in organs. However, the present study identified a subset of fibroblasts and smooth muscle cells which are the major constituents of organ stroma neither proliferative nor senescent in tissues of the elderly, which we termed "mid-old status" cells. Upregulation of pro-inflammatory genes (IL1B and SAA1) and downregulation of anti-inflammatory genes (SLIT2 and CXCL12) were detected in mid-old cells. In the stroma, SAA1 promotes development of the inflammatory microenvironment via upregulation of MMP9, which decreases the stability of epithelial cells present on the basement membrane, decreasing epithelial cell function. Remarkably, the microenvironmental change and the functional decline of mid-old cells could be reversed by a young cell-originated protein, SLIT2. Our data identify functional reversion of mid-old cells as a potential method to prevent or ameliorate aspects of aging-related tissue dysfunction.


Subject(s)
Aging , Cellular Senescence , Humans , Aged , Cellular Senescence/genetics , Aging/genetics , Epithelial Cells/physiology , Fibroblasts , Myocytes, Smooth Muscle
3.
Clin Lab ; 69(3)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36912312

ABSTRACT

BACKGROUND: We established reference intervals for research parameters of complete blood cell count and examined their usefulness for diagnosing certain diseases. METHODS: Reference intervals for 26 basic and 38 research parameters were established for 3,457 and 1,325 men and 2,742 and 830 women aged 20 - 59 and ≥ 60 years, respectively. Research parameter values for patients with iron deficiency anemia (IDA), appendicitis, sepsis, and myelodysplastic syndromes (MDS) were compared against gender- and age-matched reference values. RESULTS: Seven basic and 10 research parameters among men and one research parameter among women required partitioning by age. No partitioning by gender was required. Further, 67% patients with IDA showed micro red blood cell ratio values above the upper reference limits of their corresponding age and gender subgroups; 3% and 5% patients with appendicitis showed immature granulocyte percentages and counts above the upper reference limits, respectively; 12% - 42% of patients with sepsis showed numerous values exceeded their reference limits, and 67% and 100% patients with MDS showed neutrophil cell complexity and structural dispersion values outside their reference ranges, respectively. CONCLUSIONS: Overall, < 60% of research parameter values were outside their reference ranges among most patients, indicating their limited diagnostic usefulness.


Subject(s)
Anemia, Iron-Deficiency , Appendicitis , Hematology , Myelodysplastic Syndromes , Male , Humans , Female , Blood Cell Count , Granulocytes , Reference Values
4.
Heliyon ; 9(2): e13170, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36785830

ABSTRACT

Senescent tumor cells are nonproliferating tumor cells which are closely related to cancer progression by secreting senescence-related molecules, called senescence-associated secreting phenotypes. Therefore, the presence of senescent tumor cells is considered a prognostic factor in various cancer types. Although senescence-associated ß-galactosidase staining is considered the best marker for detection of senescent tumor cells, it can only be performed in fresh-frozen tissues. p16INK4A, a cyclin-dependent inhibitor, has been used as an alternative marker to detect senescent tumor cells in formalin-fixed paraffin-embedded tissues. However, other reliable markers to detect senescent tumor cells is still lacking. In the present study, using public single-cell RNA-sequencing data, we found that p15INK4B, a cyclin-dependent kinase inhibitor, is a novel marker for detection of senescent tumor cells. Moreover, p15INK4B expression was positively correlated with that of p16INK4A in colorectal cancer tissues. In in vitro studies, mRNA expression of p15INK4B was increased together with that of p16INK4A in H2O2- and therapy-induced cancer senescence models. However, the mRNA level of p15INK4B did not increase in the oncogene-induced senescence model in primary colonic epithelial cells. In conclusion, p15INK4B is a potential alternative marker for detection of senescent tumor cells together with conventional markers in advanced stages of colorectal cancer.

5.
Cells ; 11(13)2022 06 30.
Article in English | MEDLINE | ID: mdl-35805162

ABSTRACT

While mitochondrial bioenergetic deregulation has long been implicated in cellular senescence, its mechanistic involvement remains unclear. By leveraging diverse mitochondria-related gene expression profiles derived from two different cellular senescence models of human diploid fibroblasts, we found that the expression of mitoribosomal proteins (MRPs) was generally decreased during the early-to-middle transition prior to the exhibition of noticeable SA-ß-gal activity. Suppressed expression patterns of the identified senescence-associated MRP signatures (SA-MRPs) were validated in aged human cells and rat and mouse skin tissues and in aging mouse fibroblasts at single-cell resolution. TIN2- and POT1-interaction protein (TPP1) was concurrently suppressed, which induced senescence, accompanied by telomere DNA damage. Lastly, we show that SA-MRP deregulation could be a potential upstream regulator of TPP1 suppression. Our results indicate that mitoribosomal deregulation could represent an early event initiating mitochondrial dysfunction and serve as a primary driver of cellular senescence and an upstream regulator of shelterin-mediated telomere deprotection.


Subject(s)
Cellular Senescence , Mitochondria , Ribosomes , Shelterin Complex , Telomere-Binding Proteins , Animals , Cellular Senescence/physiology , Mice , Mitochondria/metabolism , Rats , Ribosomes/metabolism , Telomere/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
6.
Cell Death Dis ; 12(11): 1076, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772924

ABSTRACT

Deregulated mitochondrial energetics is a metabolic hallmark of cancer cells. However, the causative mechanism of the bioenergetic deregulation is not clear. In this study, we show that somatic copy number alteration (SCNA) of mitoribosomal protein (MRP) genes is a key mechanism of bioenergetic deregulation in hepatocellular carcinoma (HCC). Association analysis between the genomic and transcriptomic profiles of 82 MRPs using The Cancer Genome Atlas-Liver HCC database identified eight key SCNA-dependent MRPs: MRPS31, MRPL10, MRPL21, MRPL15, MRPL13, MRPL55, and DAP3. MRPS31 was the only downregulated MRP harboring a DNA copy number (DCN) loss. MRPS31 loss was associated specifically with the DCN losses of many genes on chromosome 13q. Survival analysis revealed a unique dependency of HCC on the MRPS31 deficiency, showing poor clinical outcome. Subclass prediction analysis using several public classifiers indicated that MRPS31 loss is linked to aggressive HCC phenotypes. By employing hepatoma cell lines with SCNA-dependent MRPS31 expression (JHH5, HepG2, Hep3B, and SNU449), we demonstrated that MRPS31 deficiency is the key mechanism, disturbing the whole mitoribosome assembly. MRPS31 suppression enhanced hepatoma cell invasiveness by augmenting MMP7 and COL1A1 expression. Unlike the action of MMP7 on extracellular matrix destruction, COL1A1 modulated invasiveness via the ZEB1-mediated epithelial-to-mesenchymal transition. Finally, MRPS31 expression further stratified the high COL1A1/DDR1-expressing HCC groups into high and low overall survival, indicating that MRPS31 loss is a promising prognostic marker. SIGNIFICANCE: Our results provide new mechanistic insight for mitochondrial deregulation in HCC and present MRPS31 as a novel biomarker of HCC malignancy.


Subject(s)
Autoantigens/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Genomics/methods , Liver Neoplasms/genetics , Ribosomal Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology , Transfection
7.
Adv Sci (Weinh) ; 8(4): 2002497, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33643790

ABSTRACT

Cellular senescence can either support or inhibit cancer progression. Here, it is shown that intratumoral infiltration of CD8+ T cells is negatively associated with the proportion of senescent tumor cells in colorectal cancer (CRC). Gene expression analysis reveals increased expression of C-X-C motif chemokine ligand 12 (CXCL12) and colony stimulating factor 1 (CSF1) in senescent tumor cells. Senescent tumor cells inhibit CD8+ T cell infiltration by secreting a high concentration of CXCL12, which induces a loss of CXCR4 in T cells that result in impaired directional migration. CSF1 from senescent tumor cells enhance monocyte differentiation into M2 macrophages, which inhibit CD8+ T cell activation. Neutralization of CXCL12/CSF1 increases the effect of anti-PD1 antibody in allograft tumors. Furthermore, inhibition of CXCL12 from senescent tumor cells enhances T cell infiltration and results in reducing the number and size of tumors in azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced CRC. These findings suggest senescent tumor cells generate a cytokine barrier protecting nonsenescent tumor cells from immune attack and provide a new target for overcoming the immunotherapy resistance of CRC.

8.
Cancers (Basel) ; 12(9)2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32942643

ABSTRACT

Mitochondrial respiratory defects have been implicated in cancer progression and metastasis, but how they control tumor cell aggressiveness remains unclear. Here, we demonstrate that a mitochondrial respiratory defect induces nuclear factor-erythroid 2 like 1 (NFE2L1) expression at the transcriptional level via reactive oxygen species (ROS)-mediated STAT3 activation. We identified syntaxin 12 (STX12) as an effective downstream target of NFE2L1 by performing cDNA microarray analysis after the overexpression and depletion of NFE2L1 in hepatoma cells. Bioinformatics analysis of The Cancer Genome Atlas Liver Hepatocellular carcinoma (TCGA-LIHC) open database (n = 371) also revealed a significant positive association (r = 0.3, p = 2.49 × 10-9) between NFE2L1 and STX12 expression. We further demonstrated that STX12 is upregulated through the ROS/STAT3/NFE2L1 axis and is a key downstream effector of NFE2L1 in modulating hepatoma cell invasiveness. In addition, gene enrichment analysis of TCGA-LIHC also showed that epithelial-mesenchymal transition (EMT)-related core genes are significantly upregulated in tumors co-expressing NFE2L1 and STX12. The positive association between NFE2L1 and STX12 expression was validated by immunohistochemistry of the hepatocellular carcinoma tissue array. Finally, higher EMT gene enrichment and worse overall survival (p = 0.043) were observed in the NFE2L1 and STX12 co-expression group with mitochondrial defect, as indicated by low NDUFA9 expression. Collectively, our results indicate that NFE2L1 is a key mitochondrial retrograde signaling-mediated primary gene product enhancing hepatoma cell invasiveness via STX12 expression and promoting liver cancer progression.

9.
iScience ; 23(6): 101247, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32629612

ABSTRACT

Mitochondrial ribosomes (mitoribosomes), the specialized translational machinery for mitochondrial genes, exclusively encode the subunits of the oxidative phosphorylation (OXPHOS) system. Although OXPHOS dysfunctions are associated with hepatic disorders including hepatocellular carcinoma (HCC), their underlying mechanisms remain poorly elucidated. In this study, we aimed to investigate the effects of mitoribosome defects on OXPHOS and HCC progression. By generating a gene signature from HCC transcriptome data, we developed a scoring system, i.e., mitoribosome defect score (MDS), which represents the degree of mitoribosomal defects in cancers. The MDS showed close associations with the clinical outcomes of patients with HCC and with gene functions such as oxidative phosphorylation, cell-cycle activation, and epithelial-mesenchymal transition. By analyzing immune profiles, we observed that mitoribosomal defects are also associated with immunosuppression and evasion. Taken together, our results provide new insights into the roles of mitoribosome defects in HCC progression.

10.
J Biol Chem ; 294(19): 7810-7820, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30923124

ABSTRACT

Aerobic glycolysis and mitochondrial dysfunction are key metabolic features of cancer cells, but their interplay during cancer development remains unclear. We previously reported that human hepatoma cells with mitochondrial defects exhibit down-regulated lactate dehydrogenase subunit B (LDHB) expression. Here, using several molecular and biochemical assays and informatics analyses, we investigated how LDHB suppression regulates mitochondrial respiratory activity and contributes to liver cancer progression. We found that transcriptional LDHB down-regulation is an upstream event during suppressed oxidative phosphorylation. We also observed that LDHB knockdown increases inhibitory phosphorylation of pyruvate dehydrogenase (PDH) via lactate-mediated PDH kinase (PDK) activation and thereby attenuates oxidative phosphorylation activity. Interestingly, monocarboxylate transporter 1 was the major lactate transporter in hepatoma cells, and its expression was essential for PDH phosphorylation by modulating intracellular lactate levels. Finally, bioinformatics analysis of the hepatocellular carcinoma cohort from The Cancer Genome Atlas revealed that a low LDHB/LDHA ratio is statistically significantly associated with poor prognostic outcomes. A low ratio was also associated with a significant enrichment in glycolysis genes and negatively correlated with PDK1 and 2 expression, supporting a close link between LDHB suppression and the PDK-PDH axis. These results suggest that LDHB suppression is a key mechanism that enhances glycolysis and is critically involved in the maintenance and propagation of mitochondrial dysfunction via lactate release in liver cancer progression.


Subject(s)
Acidosis, Lactic/enzymology , Carcinoma, Hepatocellular/enzymology , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lactate Dehydrogenases/biosynthesis , Liver Neoplasms/enzymology , Mitochondria, Liver/enzymology , Neoplasm Proteins/blood , Oxidative Phosphorylation , Acidosis, Lactic/genetics , Acidosis, Lactic/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Lactate Dehydrogenases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Neoplasm Proteins/genetics
11.
BMB Rep ; 52(1): 5-12, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30526768

ABSTRACT

Organismal aging is accompanied by a host of progressive metabolic alterations and an accumulation of senescent cells, along with functional decline and the appearance of multiple diseases. This implies that the metabolic features of cell senescence may contribute to the organism's metabolic changes and be closely linked to age-associated diseases, especially metabolic syndromes. However, there is no clear understanding of senescent metabolic characteristics. Here, we review key metabolic features and regulators of cellular senescence, focusing on mitochondrial dysfunction and anabolic deregulation, and their link to other senescence phenotypes and aging. We further discuss the mechanistic involvement of the metabolic regulators mTOR, AMPK, and GSK3, proposing them as key metabolic switches for modulating senescence. [BMB Reports 2019; 52(1): 5-12].


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Adenylate Kinase , Animals , Cellular Senescence/genetics , Glycogen Synthase Kinase 3 , Humans , Mitochondria/physiology , Phenotype , Signal Transduction , TOR Serine-Threonine Kinases
12.
BMB Rep ; 51(6): 296-301, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29580374

ABSTRACT

Mitochondrial DNA (mtDNA) mutations are often observed in various cancer types. Although the correlation between mitochondrial dysfunction and cancer malignancy has been demonstrated by several studies, further research is required to elucidate the molecular mechanisms underlying accelerated tumor development and progression due to mitochondrial mutations. We generated an mtDNA-depleted cell line, ρ°, via long-term ethidium bromide treatment to define the molecular mechanisms of tumor malignancy induced by mitochondrial dysfunction. Mitochondrial dysfunction in ρ° cells reduced drug-induced cell death and decreased the expression of pro-apoptotic proteins including p53. The p53 expression was reduced by activation of nuclear factor-κB that depended on elevated levels of free calcium in HCT116/ρ° cells. Overall, these data provide a novel mechanism for tumor development and drug resistance due to mitochondrial dysfunction. [BMB Reports 2018; 51(6): 296-301].


Subject(s)
Calcium/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Mitochondria/genetics , Mitochondria/metabolism , NF-kappa B/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Calcium Signaling , Colorectal Neoplasms/pathology , DNA, Mitochondrial/genetics , Genes, p53 , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/physiology , Signal Transduction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
13.
J Biol Chem ; 292(49): 20208-20217, 2017 12 08.
Article in English | MEDLINE | ID: mdl-28978646

ABSTRACT

Impaired mitochondrial oxidative phosphorylation (OXPHOS) capacity, accompanied by enhanced glycolysis, is a key metabolic feature of cancer cells, but its underlying mechanism remains unclear. Previously, we reported that human hepatoma cells that harbor OXPHOS defects exhibit high tumor cell invasiveness via elevated claudin-1 (CLN1). In the present study, we show that OXPHOS-defective hepatoma cells (SNU354 and SNU423 cell lines) exhibit reduced expression of mitochondrial ribosomal protein L13 (MRPL13), a mitochondrial ribosome (mitoribosome) subunit, suggesting a ribosomal defect. Specific inhibition of mitoribosomal translation by doxycycline, chloramphenicol, or siRNA-mediated MRPL13 knockdown decreased mitochondrial protein expression, reduced oxygen consumption rate, and increased CLN1-mediated tumor cell invasiveness in SNU387 cells, which have active mitochondria. Interestingly, we also found that exogenous lactate treatment suppressed MRPL13 expression and oxygen consumption rate and induced CLN1 expression. A bioinformatic analysis of the open RNA-Seq database from The Cancer Genome Atlas (TCGA) liver hepatocellular carcinoma (LIHC) cohort revealed a significant negative correlation between MRPL13 and CLN1 expression. Moreover, in patients with low MRPL13 expression, two oxidative metabolic indicators, pyruvate dehydrogenase B expression and the ratio of lactate dehydrogenase type B to type A, significantly and negatively correlated with CLN1 expression, indicating that the combination of elevated glycolysis and deficient MRPL13 activity was closely linked to CLN1-mediated tumor activity in LIHC. These results suggest that OXPHOS defects may be initiated and propagated by lactate-mediated mitoribosomal deficiencies and that these deficiencies are critically involved in LIHC development.


Subject(s)
Carcinoma, Hepatocellular/pathology , Lactic Acid/pharmacology , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Oxidative Phosphorylation , Ribosomal Proteins/metabolism , Thiolester Hydrolases/metabolism , Carcinoma, Hepatocellular/ultrastructure , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Oxidative Phosphorylation/drug effects , Oxygen Consumption , Ribosomes/drug effects , Ribosomes/pathology
14.
J Biol Chem ; 292(9): 3729-3739, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28100769

ABSTRACT

As senescence develops, cells sequentially acquire diverse senescent phenotypes along with simultaneous multistage gene reprogramming. It remains unclear what acts as the key regulator of the collective changes in gene expression at initiation of senescent reprogramming. Here we analyzed time series gene expression profiles obtained in two different senescence models in human diploid fibroblasts: replicative senescence and H2O2-induced senescence. Our results demonstrate that suppression of DNA methyltransferase 1 (DNMT1)-mediated DNA methylation activity was an initial event prior to the display of senescent phenotypes. We identified seven DNMT1-interacting proteins, ubiquitin-like with PHD and ring finger domains 1 (UHRF1), EZH2, CHEK1, SUV39H1, CBX5, PARP1, and HELLS (also known as LSH (lymphoid-specific helicase) 1), as being commonly down-regulated at the same time point as DNMT1 in both senescence models. Knockdown experiments revealed that, among the DNMT1-interacting proteins, only UHRF1 knockdown suppressed DNMT1 transcription. However, UHRF1 overexpression alone did not induce DNMT1 expression, indicating that UHRF1 was essential but not sufficient for DNMT1 transcription. Although UHRF1 knockdown effectively induced senescence, this was significantly attenuated by DNMT1 overexpression, clearly implicating the UHRF1/DNMT1 axis in senescence. Bioinformatics analysis further identified WNT5A as a downstream effector of UHRF1/DNMT1-mediated senescence. Senescence-associated hypomethylation was found at base pairs -1569 to -1363 from the transcription start site of the WNT5A gene in senescent human diploid fibroblasts. As expected, WNT5A overexpression induced senescent phenotypes. Overall, our results indicate that decreased UHRF1 expression is a key initial event in the suppression of DNMT1-mediated DNA methylation and in the consequent induction of senescence via increasing WNT5A expression.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cellular Senescence , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Chromobox Protein Homolog 5 , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Histones/metabolism , Humans , Hydrogen Peroxide/chemistry , Male , Oligonucleotide Array Sequence Analysis , Phenotype , Promoter Regions, Genetic , Protein Binding , Protein Domains , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases , Wnt-5a Protein/metabolism , beta-Galactosidase/metabolism
16.
Vasc Specialist Int ; 32(4): 166-174, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28042556

ABSTRACT

PURPOSE: Deep venous thrombosis (DVT) and pulmonary embolism (PE) are considered as similar disease entities representing different clinical manifestations. The objectives of this study were: 1) to determine the prevalence and outcome of DVT in patients with PE; 2) to identify additional risk factors for PE-related unfavorable outcome and 30-day all-cause mortality; and 3) to establish the clinical importance of screening for concomitant DVT. MATERIALS AND METHODS: From January 2013 to December 2015, a total of 141 patients with confirmed PE were evaluated. The prevalence and outcome of DVT in patients with PE was determined. Furthermore, the potential risk factors for PE-related unfavorable outcome and 30-day all-cause mortality were also analyzed. RESULTS: The prevalence of concomitant DVT was 45.4%. PE-related unfavorable outcome was observed in 21.9% of all concomitant DVT, with all-cause mortality of 21.9%. There was no significant relationship between the presence of concomitant DVT and the development of PE-related unfavorable outcome or all-cause mortality. Our results indicated that heart rate >100/min and peripheral oxygen saturation <90% were independent predictors for PE-related unfavorable outcome. Regarding all-cause mortality, active malignancy and hypotension or shock were significant risk factors. CONCLUSION: Our findings demonstrate that approximately half of patients with PE possess DVT. However, this study failed to establish any clinical significance of concomitant DVT for PE-related unfavorable outcome and all-cause mortality. Tachycardia and hypoxemia were identified as significant predictors for PE-related unfavorable outcome along with active malignancy and hypotension or shock as significant risk factors of all-cause mortality.

17.
BMB Rep ; 48(11): 597-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26350749

ABSTRACT

Mitochondrial respiratory defect is a key bioenergetics feature of hepatocellular carcinoma (HCC) cells. However, their involvement and roles in HCC development and progression remain unclear. Recently, we identified 10 common mitochondrial defect (CMD) signature genes that may be induced by retrograde signaling-mediated transcriptional reprogramming in response to HCC mitochondrial defects. HCC patients with enriched expression of these genes had poor prognostic outcomes, such as shorter periods of overall survival and recurrence-free survival. Nuclear protein 1 (NUPR1), a key transcription regulator, was up-regulated by Ca++-mediated retrograde signaling. NUPR1-centric network analysis and a biochemical promoter-binding assay demonstrated that granulin (GRN) is a key downstream effector of NUPR1 for the regulation of HCC cell invasiveness; association analysis of the NUPR1-GRN pathway supported this conclusion. Mitochondrial respiratory defects and retrograde signaling thus play pivotal roles in HCC progression, highlighting the potential of the NUPR1-GRN axis as a novel diagnostic marker and therapeutic target for HCC.


Subject(s)
Liver Neoplasms/genetics , Mitochondria, Liver/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Disease Progression , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Mitochondria, Liver/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prognosis , Progranulins , Signal Transduction , Transcriptome
18.
Hepatology ; 62(4): 1174-89, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26173068

ABSTRACT

UNLABELLED: Many cancer cells require more glycolytic adenosine triphosphate production due to a mitochondrial respiratory defect. However, the roles of mitochondrial defects in cancer development and progression remain unclear. To address the role of transcriptomic regulation by mitochondrial defects in liver cancer cells, we performed gene expression profiling for three different cell models of mitochondrial defects: cells with chemical respiratory inhibition (rotenone, thenoyltrifluoroacetone, antimycin A, and oligomycin), cells with mitochondrial DNA depletion (Rho0), and liver cancer cells harboring mitochondrial defects (SNU354 and SNU423). By comparing gene expression in the three models, we identified 10 common mitochondrial defect-related genes that may be responsible for retrograde signaling from cancer cell mitochondria to the intracellular transcriptome. The concomitant expression of the 10 common mitochondrial defect genes is significantly associated with poor prognostic outcomes in liver cancers, suggesting their functional and clinical relevance. Among the common mitochondrial defect genes, we found that nuclear protein 1 (NUPR1) is one of the key transcription regulators. Knockdown of NUPR1 suppressed liver cancer cell invasion, which was mediated in a Ca(2+) signaling-dependent manner. In addition, by performing an NUPR1-centric network analysis and promoter binding assay, granulin was identified as a key downstream effector of NUPR1. We also report association of the NUPR1-granulin pathway with mitochondrial defect-derived glycolytic activation in human liver cancer. CONCLUSION: Mitochondrial respiratory defects and subsequent retrograde signaling, particularly the NUPR1-granulin pathway, play pivotal roles in liver cancer progression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , Mitochondria/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Carcinoma, Hepatocellular/pathology , Disease Progression , Humans , Liver Neoplasms/pathology , Tumor Cells, Cultured
19.
J Biol Chem ; 290(35): 21421-31, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26157141

ABSTRACT

Although mitochondrial dysfunction has been implicated in tumor metastasis, it is unclear how it regulates tumor cell aggressiveness. We have reported previously that human hepatoma cells harboring mitochondrial defects have high tumor cell invasion activity via increased claudin-1 (Cln-1) expression. In this study, we demonstrated that mitochondrial respiratory defects induced Cln-1 transcription via reactive oxygen species (ROS)-mediated heat shock factor 1 (HSF1) activation, which contributed to hepatoma invasiveness. We first confirmed the inverse relationship between mitochondrial defects and Cln-1 induction in SNU hepatoma cells and hepatocellular carcinoma tissues. We then examined five different respiratory complex inhibitors, and complex I inhibition by rotenone most effectively induced Cln-1 at the transcriptional level. Rotenone increased both mitochondrial and cytosolic ROS. In addition, rotenone-induced Cln-1 expression was attenuated by N-acetylcysteine, an antioxidant, and exogenous H2O2 treatment was enough to increase Cln-1 transcription, implying the involvement of ROS. Next we found that ROS-mediated HSF1 activation via hyperphosphorylation was the key event for Cln-1 transcription. Moreover, the Cln-1 promoter region (from -529 to +53) possesses several HSF1 binding elements, and this region showed increased promoter activity and HSF1 binding affinity in response to rotenone treatment. Finally, we demonstrated that the invasion activity of SNU449 cells, which harbor mitochondrial defects, was blocked by siRNA-mediated HSF1 knockdown. Taken together, these results indicate that mitochondrial respiratory defects enhance Cln-1-mediated hepatoma cell invasiveness via mitochondrial ROS-mediated HSF1 activation, presenting a potential role for HSF1 as a novel mitochondrial retrograde signal-responsive transcription factor to control hepatoma cell invasiveness.


Subject(s)
Carcinoma, Hepatocellular/pathology , Claudin-1/genetics , DNA-Binding Proteins/metabolism , Liver Neoplasms/pathology , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Base Sequence , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Respiration , Claudin-1/metabolism , Gene Expression Regulation, Neoplastic , Heat Shock Transcription Factors , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mitochondria/metabolism , Molecular Sequence Data , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic
20.
BMB Rep ; 48(10): 549-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26129674

ABSTRACT

Cellular senescence is a process by which cells enter a state of permanent cell cycle arrest. It is commonly believed to underlie organismal aging and age-associated diseases. However, the mechanism by which cellular senescence contributes to aging and age-associated pathologies remains unclear. Recent studies showed that senescent cells exert detrimental effects on the tissue microenvironment, generating pathological facilitators or aggravators. The most significant environmental effector resulting from senescent cells is the senescence-associated secretory phenotype (SASP), which is constituted by a strikingly increased expression and secretion of diverse pro-inflammatory cytokines. Careful investigation into the components of SASPs and their mechanism of action, may improve our understanding of the pathological backgrounds of age-associated diseases. In this review, we focus on the differential expression of SASP-related genes, in addition to SASP components, during the progress of senescence. We also provide a perspective on the possible action mechanisms of SASP components, and potential contributions of SASP-expressing senescent cells, to age-associated pathologies.


Subject(s)
Cellular Senescence/physiology , Age Factors , Animals , Cellular Senescence/genetics , Humans , Phenotype , Secretory Pathway/genetics , Secretory Pathway/physiology
SELECTION OF CITATIONS
SEARCH DETAIL