Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Exp Pharmacol Physiol ; 51(1): 3-9, 2024 01.
Article in English | MEDLINE | ID: mdl-37840030

ABSTRACT

Children, as a special group, have their own peculiarities in terms of individualized medication use compared to adults. Adverse drug reactions have been an important issue that needs to be addressed in the hope of safe medication use in children, and the occurrence of adverse drug reactions is partly due to genetic factors. Anti-infective drugs are widely used in children, and they have always been an important cause of the occurrence of adverse reactions in children. Pharmacogenomic technologies are becoming increasingly sophisticated, and there are now many guidelines describing the pharmacogenomics of anti-infective drugs. However, data from paediatric-based studies are scarce. This review provides a systematic review of the pharmacogenomics of anti-infective drugs recommended for gene-guided use in CPIC guidelines by exploring the relationship between pharmacogenetic frequencies and the incidence of adverse reactions, which will help inform future studies of individualized medication use in children.


Subject(s)
Anti-Infective Agents , Drug-Related Side Effects and Adverse Reactions , Adult , Humans , Child , Pharmacogenetics , Drug-Related Side Effects and Adverse Reactions/genetics , Anti-Infective Agents/adverse effects
2.
Int J Mol Sci ; 25(1)2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38203794

ABSTRACT

Stabilization of a G-quadruplex (G4) in the promotor of the c-MYC proto-oncogene leads to inhibition of gene expression, and it thus represents a potentially attractive new strategy for cancer treatment. However, most G4 stabilizers show little selectivity among the many G4s present in the cellular complement of DNA and RNA. Intriguingly, a crescent-shaped cell-penetrating thiazole peptide, TH3, preferentially stabilizes the c-MYC G4 over other promotor G4s, but the mechanisms leading to this selective binding remain obscure. To investigate these mechanisms at the atomic level, we performed an in silico comparative investigation of the binding of TH3 and its analogue TH1 to the G4s from the promotors of c-MYC, c-KIT1, c-KIT2, and BCL2. Molecular docking and molecular dynamics simulations, combined with in-depth analyses of non-covalent interactions and bulk and per-nucleotide binding free energies, revealed that both TH3 and TH1 can induce the formation of a sandwich-like framework through stacking with both the top and bottom G-tetrads of the c-MYC G4 and the adjacent terminal capping nucleotides. This framework produces enhanced binding affinities for c-MYC G4 relative to other promotor G4s, with TH3 exhibiting an outstanding binding priority. Van der Waals interactions were identified to be the key factor in complex formation in all cases. Collectively, our findings fully agree with available experimental data. Therefore, the identified mechanisms leading to specific binding of TH3 towards c-MYC G4 provide valuable information to guide the development of new selective G4 stabilizers.


Subject(s)
Genes, myc , Molecular Docking Simulation , Peptides/pharmacology , Thiazoles/pharmacology
3.
Clin Exp Pharmacol Physiol ; 48(9): 1185-1202, 2021 09.
Article in English | MEDLINE | ID: mdl-34046925

ABSTRACT

Cancer stem cells (CSCs) are a small population of heterogeneous tumor cells with the capacity of self-renewal and aberrant differentiation for immortality and divergent lineages of cancer cells. In contrast to bulky tumor cells, CSCs remain less differentiated and resistant to therapy even when targeted with tissue-specific antigenic markers. This makes CSCs responsible for not only tumor initiation, development, but also tumor recurrence. Emerging evidence suggests that CSCs can undergo cell senescence, a non-proliferative state of cells in response to stress. While cell senescence attenuates tumor cell proliferation, it is commonly regarded as a tumor suppressive mechanism. However, mounting research indicates that CSC senescence also provides these cells with the capacity to evade cytotoxic effects from cancer therapy, exacerbating cancer relapse and metastasis. Recent studies demonstrate that senescence drives reprogramming of cancer cell toward stemness and promotes CSC generation. In this review, we highlight the origin, heterogeneity and senescence regulatory mechanisms of CSCs, the complex relationship between CSC senescence and tumor therapy, and the recent beneficial effects of senotherapy on eliminating senescent tumor cells.


Subject(s)
Neoplastic Stem Cells
4.
Clin Exp Pharmacol Physiol ; 48(5): 651-659, 2021 05.
Article in English | MEDLINE | ID: mdl-33634502

ABSTRACT

Pulmonary senescence and fibrosis occur with deoxyribonucleic acid (DNA) damage response in the lungs deficient of telomerase. The molecular mechanism mediating pulmonary alveolar cell fates remains to be investigated. The present study shows that pulmonary alveolar epithelial type 2 cells (AEC2) (alveolar stem cells) undergo not only replicative senescence, but also apoptosis and differentiation in association with increased innate immune natural killer (NK) cells in telomerase knockout (KO) mice. Telomerase ribonucleic acid (RNA) component (TERC) deficiency results in increased senescence-associated heterochromatin foci marker HP1γ, p21, p16 and apoptosis-associated cleaved caspase-3 in AEC2. However, p53 deficiency in the Trp53-/- allele of the late generation of TERC KO mice attenuates the increased senescent and apoptotic markers significantly. Moreover, p53 deficiency has no significant effect on the increased gene expression of T1α (a marker of terminal differentiated alveolar epithelial type 1 cells [AEC1]) in AEC2 of the late generation of TERC KO mice. Collectively, our findings suggest that pulmonary senescence takes place in deficiency of telomerase RNA component with the alveolar stem cells undergoing p53-dependent senescence and apoptosis as well as p53-independent differentiation.


Subject(s)
Telomerase , Alveolar Epithelial Cells , Animals , Mice , Tumor Suppressor Protein p53
5.
Genes Chromosomes Cancer ; 59(2): 73-83, 2020 02.
Article in English | MEDLINE | ID: mdl-31408253

ABSTRACT

Hypoxia-induced epithelial-mesenchymal transition (EMT) involves the interplay between chromatin modifiers histone deacetylase 3 (HDAC3) and WDR5. The histone mark histone 3 lysine 4 acetylation (H3K4Ac) is observed in the promoter regions of various EMT marker genes (eg, CDH1 and VIM). To further define the genome-wide location of H3K4Ac, a chromatin immunoprecipitation followed by massively parallel DNA sequencing (ChIP-seq) analysis was performed using a head and neck squamous cell carcinoma (HNSCC) FaDu cell line under normoxia and hypoxia. H3K4Ac was found to be located mainly around the transcription start site. Coupled with analysis of gene expression by RNA sequencing and using a HDAC3 knockdown cell line, 10 new genes (BMI1, GLI1, SMO, FOXF1, SIRT2, etc) that were labeled by H3K4Ac and regulated by HDAC3 were identified. Overexpression or knockdown of GLI1/SMO increased or repressed the in vitro migration and invasion activity in OECM-1/FaDu cells, respectively. In HNSCC patients, coexpression of GLI1 and SMO in primary tumors correlated with metastasis. Our results identify new EMT marker genes that may play a significant role in hypoxia-induced EMT and metastasis and further provide diagnostic and prognostic implications.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Histone Deacetylases/genetics , Histones/genetics , Acetylation , Antigens, CD/genetics , Cadherins/genetics , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , High-Throughput Nucleotide Sequencing/methods , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism
6.
Clin Gastroenterol Hepatol ; 18(11): 2564-2572.e1, 2020 10.
Article in English | MEDLINE | ID: mdl-32109631

ABSTRACT

BACKGROUND & AIMS: Portal vein thrombosis (PVT) is a common and serious complication in patients with cirrhosis. However, little is known about PVT in patients with cirrhosis and acute decompensation (AD). We investigated the prevalence and clinical significance of PVT in nonmalignant patients with cirrhosis and AD. METHODS: We performed a retrospective study of 2 cohorts of patients with acute exacerbation of chronic liver disease who participated in the Chinese AcuTe on CHronic LIver FailurE study, established by the Chinese Chronic Liver Failure Consortium, from January 2015 through December 2016 (n = 2600 patients) and July 2018 through January 2019 (n = 1370 patients). We analyzed data on the prevalence, clinical manifestations, and risk factors of PVT from 2826 patients with cirrhosis, with and without AD. RESULTS: The prevalence of PVT in patients with cirrhosis and AD was 9.36%, which was significantly higher than in patients with cirrhosis without AD (5.24%) (P = .04). Among patients with cirrhosis and AD, 63.37% developed PVT recently (the first detected PVT with no indication of chronic PVT). Compared with patients without PVT, a significantly higher proportion of patients with PVT had variceal bleeding (47.33% vs 19.63%; P < .001) and patients with PVT had a significantly higher median serum level of D-dimer (2.07 vs 1.25; P < .001). Splenectomy and endoscopic sclerotherapy were independent risk factors for PVT in patients with cirrhosis and AD. The 1-year mortality rate did not differ significantly between patients with vs without PVT. CONCLUSIONS: In an analysis of data from 2826 patients with cirrhosis, a significantly higher proportion of those with AD had PVT than those without AD. PVT was associated with increased variceal bleeding, which would increase the risk for AD. Strategies are needed to prevent PVT in patients with cirrhosis, through regular screening, to reduce portal hypertension. ClinicalTrials.gov no: NCT02457637 and NCT03641872.


Subject(s)
Esophageal and Gastric Varices , Venous Thrombosis , Esophageal and Gastric Varices/complications , Esophageal and Gastric Varices/epidemiology , Esophageal and Gastric Varices/pathology , Gastrointestinal Hemorrhage/pathology , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Portal Vein/pathology , Prevalence , Retrospective Studies , Venous Thrombosis/complications , Venous Thrombosis/epidemiology , Venous Thrombosis/pathology
7.
BMC Med ; 18(1): 250, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32762696

ABSTRACT

BACKGROUND: COVID-19 is an extremely severe infectious disease. However, few studies have focused on the epidemiological and clinical characteristics of pediatric COVID-19. This study conducted a retrospective review of the epidemiological and clinical features of COVID-19 in children. METHODS: A retrospective study was conducted on children with a definite diagnosis of COVID-19 in mainland China using the web crawler technique to collect anonymous COVID-19 updates published by local health authorities. RESULTS: Three hundred forty-one children aged 4 days to 14 years with a median age of 7 years were included. Sixty-six percent of pediatric patients were infected via family members with COVID-19. The median incubation period was 9 days (interquartile range, 6 to 13). Asymptomatic cases accounted for 5.9%, of which 30% had abnormal chest radiologic findings. A majority of pediatric COVID-19 cases showed mild to moderate clinical features, and only a few developed severe or critical diseases (0.6% and 0.3%, respectively). Fever (77.9%) and cough (32.4%) were the predominant presenting symptoms of pediatric COVID-19. The pediatric patients had fewer underlying diseases and complications than adults. The treatment modalities for pediatric COVID-19 patients were not as complex as those of adult COVID-19 patients. The overall prognosis of pediatric COVID-19 was benign with a decent recovery. The median time from onset to cure was 16 days (interquartile range, 13 to 21). CONCLUSIONS: Compared to adults, COVID-19 in children has distinct features of epidemiology and clinical manifestations. The findings from this study might help to guide the development of measures to prevent and treat this ongoing global pandemic. TRIAL REGISTRATION: Chinese Clinical Trial Registry ( chictr.org.cn ) identifier: ChiCTR2000030464.


Subject(s)
Betacoronavirus , Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , Adolescent , COVID-19 , Child , Child, Preschool , China/epidemiology , Cough/etiology , Female , Fever/etiology , Humans , Infant , Infant, Newborn , Male , Pandemics , Retrospective Studies , SARS-CoV-2
8.
Yeast ; 37(7-8): 373-388, 2020 07.
Article in English | MEDLINE | ID: mdl-32639041

ABSTRACT

Subtelomere Y' elements get amplified by homologous recombination in sustaining the survival and division of the budding yeast Saccharomyces cerevisiae. However, current method for measurement of the subtelomere structures uses Southern blotting with labelled specific probes, which is laborious and time-consuming. By multiple sequence alignment analysis of all 19 subtelomere Y' elements across the 13 chromosomes of the sequenced S288C strain deposited in the yeast genome SGD database, we identified 12 consensus and relative longer fragments and 14 pairs of unique primers for real-time quantitative PCR analysis. With a PAC2 or ACT1 located near the centromere of chromosome V and VI as internal controls, these primers were applied to real-time quantitative PCR analysis, so the relative Y' element intensity normalised to that of wild type (WT) cells was calculated for subtelomere Y' element copy numbers across all different chromosomes using the formula: 2^[-((CTmutant Y' - CTmutant control ) - (CTWT Y' - CTWT control ))]. This novel quantitative subtelomere amplification assay across chromosomes by real-time PCR proves to be a much simpler and more sensitive way than the traditional Southern blotting method to analyse the Y' element recombination events in survivors derived from telomerase deficiency or recruitment failure.


Subject(s)
Chromosomes , Genomics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Actins/genetics , Base Sequence , DNA, Fungal/genetics , Microtubule-Associated Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomerase/genetics , Telomerase/metabolism
9.
Yeast ; 37(11): 585-595, 2020 11.
Article in English | MEDLINE | ID: mdl-32776370

ABSTRACT

Telomere length is measured using Southern blotting of the chromosomal terminal restriction fragments (TRFs) released by endonuclease digestion in cells from yeast to human. In the budding yeast Saccharomyces cerevisiae, XhoI or PstI is applied to cut the subtelomere Y' element and release TRFs from the 17 subtelomeres. However, telomeres from other 15 X-element-only subtelomeres are omitted from analysis. Here, we report a method for measuring all 32 telomeres in S. cerevisiae using the endonuclease MmeI. Based on analyses of the endonuclease cleavage sites, we found that the TRFs generated by MmeI displayed two distinguishable bands in the sizes of ~500 and ~700 bp comprising telomeres (300 bp) and subtelomeres (200-400 bp). The modified MmeI-restricted TRF (mTRF) method recapitulated telomere shortening and lengthening caused by deficiencies of YKu and Rif1 respectively in S. cerevisiae. Furthermore, we found that mTRF was also applicable to telomere length analysis in S. paradoxus strains. These results demonstrate a useful tool for simultaneous detection of telomeres from all chromosomal ends with both X-element-only and Y'-element subtelomeres in S. cerevisiae species.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Telomere/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Telomere/genetics , Telomere Shortening , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
10.
Clin Exp Pharmacol Physiol ; 47(3): 357-364, 2020 03.
Article in English | MEDLINE | ID: mdl-31799699

ABSTRACT

Telomeres are specialized genomic structures that protect chromosomal ends to maintain genomic stability. Telomeric length is primarily regulated by the telomerase complex, essentially consisting of an RNA template (TERC), an enzymatic subunit (telomerase reverse transcriptase, TERT). In humans, telomerase activity is repressed during embryonic differentiation and is absent in most somatic cells. However, it is upregulated or reactivated in 80%-90% of the primary tumours in humans. The human TERT (hTERT) plays a pivotal role in cellular immortality and tumourigenesis. However, the molecular mechanisms of telomerase functioning in cancer have not been fully understood beyond the telomere maintenance. Several research groups, including ours, have demonstrated that hTERT possesses vital functions independent of its telomere maintenance, including angiogenesis, inflammation, cancer cell stemness, and epithelial-mesenchymal transformation (EMT). All these telomere-independent activities of hTERT may contribute to the regulation of the dynamics and homeostasis of the tumour microenvironment (TME), thereby promoting tumour growth and development. Cancer progression and metastasis largely depend upon the interactions between cancer cells and their microenvironment. In this review, the involvement of TERT in the tumour microenvironment and the underlying implications in cancer therapeutics have been summarized.


Subject(s)
Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Telomerase/metabolism , Tumor Microenvironment/physiology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Neoplasm Invasiveness/pathology , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Telomerase/physiology
11.
Clin Exp Pharmacol Physiol ; 47(5): 892-902, 2020 05.
Article in English | MEDLINE | ID: mdl-31894867

ABSTRACT

The single-strand human telomere overhang forms intramolecular high-order structures named G-quadruplex (G4) under physiological conditions. Telomere G4 stabilization prevents telomere lengthening by telomerase in cancer cells representing a promising strategy in cancer therapy. Using molecular docking and molecular dynamics (MD) simulations, specific binding of the anionic phthalocyanine 3,4',4'',4'''-tetrasulfonic acid (APC) to the human hybrid (3 + 1) G4s was investigated at the atomic level. We found that APC preferred the end-stacking binding with the telomere hybrid type II (hybrid-II) G4 as compared to the groove binding with the hybrid type I (hybrid-I) G4 remarkable stabilizing effect and more favourable binding free energies. Analysis of non-covalent interaction and decomposition of the binding free energy revealed that van der Waals interaction played a leading role in the binding of APC and telomere hybrid G4s. These findings provide evidence for the first time to shed light on the designs of selective telomere G4 stabilizers.


Subject(s)
G-Quadruplexes , Indoles/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Telomere/metabolism , Binding Sites , Humans , Indoles/pharmacology , Isoindoles , Ligands , Nucleic Acid Conformation , Telomere/genetics
12.
Clin Exp Pharmacol Physiol ; 46(2): 105-115, 2019 02.
Article in English | MEDLINE | ID: mdl-30565707

ABSTRACT

Recent clinical success of immunotherapy that inhibits the negative immune regulatory pathway programmed cell death protein-1/PD-1 ligand 1 (PD-1/PD-L1) has initiated a new era in the treatment of metastatic cancer. However, greater challenges remain to treat all cancers. The molecular architecture in the immune synapse constituting positive engagements for immune activation and negative checkpoints against immune hyperactivity is regulated dynamically by interaction between proteostasis and tumour microenvironment. This article reviews recent progresses in our understandings of the cellular and molecular mechanisms of the negative checkpoint PD-1/PD-L1 behaviours in immune tolerance of tumourigenesis and metastasis. We provide an overview on PD-L1 gene expression regulation, protein turnover, intra- and extracellular trafficking, exosome-mediated inter-cellular transport, molecular interface peptide mimetics, inhibitory chemical compounds such as metformin, and antibody dynamics. We summarise PD-L1 post-translational modifications including glycosylation, palmitoylation, phosphorylation and ubiquitination, reflecting future research directions and opportunities in identifying tumour-specific signalling targets, their regulatory molecules and pathways for intervention into various types of cancers.


Subject(s)
Antibodies, Monoclonal/immunology , Immune Tolerance , Immunotherapy/methods , Neoplasms/immunology , Peptidomimetics/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
13.
Clin Exp Pharmacol Physiol ; 46(8): 689-693, 2019 08.
Article in English | MEDLINE | ID: mdl-31009088

ABSTRACT

Genetic polymorphisms impact biological responses to drugs. Current pharmacogenomics guidelines formulated by different countries, such as the Clinical Pharmacogenetics Implementation Consortium, the Dutch Pharmacogenetics Working Group, the Canadian Pharmacogenomics Network for Drug Safety, and the French National Network (Réseau) of Pharmacogenetics, play important roles in clinical practices. However, the standards for these guidelines vary significantly, resulting in differences in recommendations. The present article discusses these differences by head-to-head comparison of the existing pharmacogenomics guidelines and proposes new strategies for their future development.


Subject(s)
Guidelines as Topic , Pharmacogenetics/methods , Humans
14.
BMC Cancer ; 18(1): 499, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29716528

ABSTRACT

BACKGROUND: PEST-containing nuclear protein (PCNP), a novel nuclear protein, is involved in cell proliferation and tumorigenesis. However, the precise mechanism of action of PCNP in the process of tumor growth has not yet been fully elucidated. METHODS: ShRNA knockdown and overexpression of PCNP were performed in human neuroblastoma cells. Tumorigenic and metastatic effects of PCNP were examined by tumor growth, migration, and invasion assays in vitro, as well as xenograft tumor assay in vivo. RESULTS: PCNP over-expression decreased the proliferation, migration, and invasion of human neuroblastoma cells and down-regulation of PCNP showed reverse effects. PCNP over-expression increased protein expressions of cleaved caspase-3, cleaved caspase-8, cleaved caspase-9, and cleaved poly adenosine diphosphate-ribose polymerase, as well as ratios of B-cell lymphoma-2 (Bcl-2)-associated X protein/Bcl-2 and Bcl-2-associated death promoter/B-cell lymphoma-extra large in human neuroblastoma cells, however PCNP knockdown exhibited reverse trends. PCNP over-expression increased phosphorylations of extracellular signal-regulated protein kinase 1/2, p38, c-Jun N-terminal kinase, as well as decreased phosphorylations of phosphatidylinositol 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR), nevertheless PCNP knockdown exhibited opposite effects. Furthermore, PCNP over-expression significantly reduced the growth of human neuroblastoma xenograft tumors by down-regulating angiogenesis, whereas PCNP knockdown markedly promoted the growth of human neuroblastoma xenograft tumors through up-regulation of angiogenesis. CONCLUSIONS: PCNP mediates the proliferation, migration, and invasion of human neuroblastoma cells through mitogen-activated protein kinase and PI3K/AKT/mTOR signaling pathways, implying that PCNP is a therapeutic target for patients with neuroblastoma.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation , Gene Expression , Gene Knockdown Techniques , Humans , Immunohistochemistry , MAP Kinase Signaling System , Male , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics
15.
Nucleic Acids Res ; 44(18): 8693-8703, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27325744

ABSTRACT

Angiogenesis is recognized as an important hallmark of cancer. Although telomerase is thought to be involved in tumor angiogenesis, the evidence and underlying mechanism remain elusive. Here, we demonstrate that human telomerase reverse transcriptase (hTERT) activates vascular epithelial growth factor (VEGF) gene expression through interactions with the VEGF promoter and the transcription factor Sp1. hTERT binds to Sp1 in vitro and in vivo and stimulates angiogenesis in a manner dependent on Sp1. Deletion of the mTert gene in the first generation of Tert null mice compromised tumor growth, with reduced VEGF expression. In addition, we show that hTERT expression levels are positively correlated with those of VEGF in human gastric tumor samples. Together, our results demonstrate that hTERT facilitates tumor angiogenesis by up-regulating VEGF expression through direct interactions with the VEGF gene and the Sp1 transcription factor. These results provide novel insights into hTERT function in tumor progression in addition to its role in telomere maintenance.


Subject(s)
Neovascularization, Pathologic/metabolism , Sp1 Transcription Factor/metabolism , Telomerase/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , HeLa Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Neovascularization, Physiologic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Promoter Regions, Genetic , Protein Binding/genetics , Stomach Neoplasms/blood supply , Stomach Neoplasms/genetics , Transcription, Genetic , Up-Regulation/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Proc Natl Acad Sci U S A ; 112(5): 1535-40, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605927

ABSTRACT

Inflammation is critical for host defense, but without appropriate control, it can cause chronic disease or even provoke fatal responses. Here we identify a mechanism that limits the inflammatory response. Probing the responses of macrophages to the key sensory Toll-like receptors, we identify that the Broad-complex, Tramtrack and Bric-a-brac/poxvirus and zinc finger (BTB/POZ), transcriptional regulator promyelocytic leukemia zinc finger (PLZF) limits the expression of inflammatory gene products. In accord with this finding, PLZF-deficient animals express higher levels of potent inflammatory cytokines and mount exaggerated inflammatory responses to infectious stimuli. Temporal quantitation of inflammatory gene transcripts shows increased gene induction in the absence of PLZF. Genome-wide analysis of histone modifications distinguish that PLZF establishes basal activity states of early response genes to maintain immune homeostasis and limit damaging inflammation. We show that PLZF stabilizes a corepressor complex that encompasses histone deacetylase activity to control chromatin. Together with our previous demonstration that PLZF promotes the antiviral response, these results suggest a strategy that could realize one of the major goals of immune therapy to retain immune resistance to pathogens while curbing damaging inflammation.


Subject(s)
Chromatin/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , Signal Transduction , Animals , Bacterial Infections/metabolism , Chromatin Immunoprecipitation , Fluorescence Resonance Energy Transfer , Histone Deacetylases/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Real-Time Polymerase Chain Reaction
17.
Clin Exp Pharmacol Physiol ; 44 Suppl 1: 3-8, 2017 12.
Article in English | MEDLINE | ID: mdl-29178613

ABSTRACT

Premature aging occurs frequently to various tissues and organs resulting in the tissue-specific chronic diseases. The mechanisms of tissue-specific premature aging are largely unknown. In response to environmental cues, aging may originate from cytoplasm or the nucleus of a cell with cytoplasm aging in association with organelle degeneration in terminally differentiated cells and nuclear aging with dysfunctional telomeres and irreversible cell cycle arrest in stem and cancer cells. Either cytoplasm aging or nuclear aging may cause extracellular senescence-associated low-grade inflammation to spread aging. Referring to the recent findings in this special issue of Healthy Aging in CEPP and beyond, we describe the molecular and cellular mechanisms of physiological aging and tissue-specific pathological aging in chronic diseases.


Subject(s)
Aging, Premature , Aging , Cellular Senescence , Signal Transduction , Age Factors , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cellular Senescence/genetics , Chronic Disease , Female , Health Status , Humans , Male , Mitochondria/metabolism , Mitochondria/pathology , Signal Transduction/genetics
18.
Clin Exp Pharmacol Physiol ; 44(2): 172-179, 2017 02.
Article in English | MEDLINE | ID: mdl-27997702

ABSTRACT

Lysosomal transport of cargos in neurons is essential for neuronal proteostasis, transmission and functional motors and behaviours. Lysosomal malfunction including storage disorders is involved in the pathogenesis of Parkinson's disease (PD). Given the unclear molecular mechanisms of diverse defects in PD phenotypes, especially behavioural deficits, this mini review explores the cellular contexts of PD impulse control disorders and the molecular aspects of lysosomal cross-membrane transports. Focuses are paid to trace metal involvements in α-synuclein assembly in Lewy bodies, the functions and molecular interactions of ATP13A2 as ATPase transporters in lysosomal membranes for cross-membrane trafficking and lysosomal homeostasis, and our current understandings of the neural circuits in ICD. Erroneously polarized distributions of cargos such as metals and lipids on each side of lysosomal membranes triggered by gene mutations and deregulated expression of ATP13A2 may thus instigate sensing protein structural changes such as aggregations, organelle degeneration, and specific neuronal ageing and death in Parkinsonism.


Subject(s)
Disruptive, Impulse Control, and Conduct Disorders/metabolism , Lysosomes/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Proton-Translocating ATPases/metabolism , Animals , Disruptive, Impulse Control, and Conduct Disorders/complications , Disruptive, Impulse Control, and Conduct Disorders/pathology , Humans , Intracellular Membranes/metabolism , Lewy Bodies/metabolism , Manganese/metabolism , Neurons/pathology , Parkinson Disease/complications , Parkinson Disease/pathology , Protein Transport , Proton-Translocating ATPases/genetics , alpha-Synuclein/metabolism
19.
Adv Exp Med Biol ; 1024: 213-224, 2017.
Article in English | MEDLINE | ID: mdl-28921472

ABSTRACT

Telomeres at the ends of chromosomes safeguard genome integrity and stability in human nucleated cells. However, telomere repeats shed off during cell proliferation and other stress responses. Our recent studies show that telomere attrition induces not only epithelial stem cell senescence but also low-grade inflammation in the lungs. The senescence-associated low-grade inflammation (SALI) is characteristic of alveolar stem cell replicative senescence, increased proinflammatory and anti-inflammatory cytokines, infiltrated immune cells, and spillover effects. To date, the mechanisms underlying SALI remain unclear. Investigations demonstrate that senescent epithelial stem cells with telomere erosion are not the source of secreted cytokines, containing no significant increase in expression of the genes coding for increased cytokines, suggesting an alternative senescence-associated secretory phenotype (A-SASP). Given that telomere loss results in significant alterations in the genomes and accumulations of the cleaved telomeric DNA in the cells and milieu externe, we conclude that telomere position effects (TPEs) on gene expression and damage-associated molecular patterns (DAMPs) in antigen presentation are involved in A-SASP and SALI in response to telomere damage in mammals.


Subject(s)
Inflammation/pathology , Telomere , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/immunology , Gene Expression Regulation/physiology , Humans , Inflammation/metabolism , Stem Cells/physiology
20.
J Biol Chem ; 290(52): 30813-29, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26518879

ABSTRACT

Mutations of human telomerase RNA component (TERC) and telomerase reverse transcriptase (TERT) are associated with a subset of lung aging diseases, but the mechanisms by which TERC and TERT participate in lung diseases remain unclear. In this report, we show that knock-out (KO) of the mouse gene Terc or Tert causes pulmonary alveolar stem cell replicative senescence, epithelial impairment, formation of alveolar sacs, and characteristic inflammatory phenotype. Deficiency in TERC or TERT causes a remarkable elevation in various proinflammatory cytokines, including IL-1, IL-6, CXCL15 (human IL-8 homolog), IL-10, TNF-α, and monocyte chemotactic protein 1 (chemokine ligand 2 (CCL2)); decrease in TGF-ß1 and TGFßRI receptor in the lungs; and spillover of IL-6 and CXCL15 into the bronchoalveolar lavage fluids. In addition to increased gene expressions of α-smooth muscle actin and collagen 1α1, suggesting myofibroblast differentiation, TERC deficiency also leads to marked cellular infiltrations of a mononuclear cell population positive for the leukocyte common antigen CD45, low-affinity Fc receptor CD16/CD32, and pattern recognition receptor CD11b in the lungs. Our data demonstrate for the first time that telomerase deficiency triggers alveolar stem cell replicative senescence-associated low-grade inflammation, thereby driving pulmonary premature aging, alveolar sac formation, and fibrotic lesion.


Subject(s)
Lung Diseases/immunology , Pulmonary Alveoli/enzymology , Stem Cells/cytology , Telomerase/deficiency , Animals , Cellular Senescence , Female , Humans , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lung/cytology , Lung/immunology , Lung Diseases/enzymology , Lung Diseases/genetics , Lung Diseases/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , RNA/genetics , Stem Cells/immunology , Telomerase/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL