Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Sci ; 115(1): 184-196, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38050344

ABSTRACT

p53 is a key tumor suppressor mutated in half of human cancers. In recent years, p53 was shown to regulate a wide variety of functions. From the transcriptome analysis of 24 tissues of irradiated mice, we identified 553 genes markedly induced by p53. Gene Ontology (GO) enrichment analysis found that the most associated biological process was innate immunity. 16S rRNA-seq analysis revealed that Akkermansia, which has anti-inflammatory properties and is involved in the regulation of intestinal barrier integrity, was decreased in p53-knockout (p53-/- ) mice after radiation. p53-/- mice were susceptible to radiation-induced GI toxicity and had a significantly shorter survival time than p53-wild-type (p53+/+ ) mice following radiation. However, administration of antibiotics resulted in a significant improvement in survival and protection against GI toxicity. Mbl2 and Lcn2, which have antimicrobial activity, were identified to be directly transactivated by p53 and secreted by liver into the circulatory system. We also found the expression of MBL2 and LCN2 was decreased in liver cancer tissues with p53 mutations compared with those without p53 mutations. These results indicate that p53 is involved in shaping the gut microbiome through its downstream targets related to the innate immune system, thus protecting the intestinal barrier.


Subject(s)
Gastrointestinal Microbiome , Immunity, Innate , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Liver Neoplasms/metabolism , Mannose-Binding Lectin/metabolism , Mice, Knockout , RNA, Ribosomal, 16S/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Neoplasia ; 20(7): 668-677, 2018 07.
Article in English | MEDLINE | ID: mdl-29802988

ABSTRACT

Recent studies in RAS wild-type (WT) metastatic colorectal cancer (mCRC) suggest that the survival benefits of therapy using anti-epidermal growth factor receptor (anti-EGFR) and anti-vascular endothelial growth factor (anti-VEGF) antibodies combined with chemotherapy are maximized when the anti-EGFR antibody is given as first-line, followed by subsequent anti-VEGF antibody therapy. We report reverse-translational research using LIM1215 xenografts of RAS WT mCRC to elucidate the biologic mechanisms underlying this clinical observation. Sequential administration of panitumumab then bevacizumab (PB) demonstrated a stronger tendency to inhibit tumor growth than bevacizumab then panitumumab (BP). Cell proliferation was reduced significantly with PB (P < .01) but not with BP based on Ki-67 index. Phosphoproteomic analysis demonstrated reduced phosphorylation of EGFR and EPHA2 with PB and BP compared with control. Western blotting showed reduced EPHA2 expression and S897-phosphorylation with PB; RSK phosphorylation was largely unaffected by PB but increased significantly with BP. In quantitative real-time PCR analyses, PB significantly reduced the expression of both lipogenic (FASN, MVD) and hypoxia-related (CA9, TGFBI) genes versus control. These results suggest that numerous mechanisms at the levels of gene expression, protein expression, and protein phosphorylation may explain the improved clinical activity of PB over BP in patients with RAS WT mCRC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Bevacizumab/pharmacology , Colorectal Neoplasms/pathology , Animals , Biomarkers , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Disease Models, Animal , ErbB Receptors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Hypoxia/metabolism , Mice , Panitumumab , Phosphorylation , Proteome , Receptor, EphA2/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem ; 26(9): 2452-2465, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29669694

ABSTRACT

We pursued serine palmitoyltransferase (SPT) inhibitors as novel cancer therapeutic agents based on a correlation between SPT inhibition and growth suppression of cancer cells. High-throughput screening and medicinal chemistry efforts led to the identification of structurally diverse SPT inhibitors 4 and 5. Both compounds potently inhibited SPT enzyme and decreased intracellular ceramide content. In addition, they suppressed cell growth of human lung adenocarcinoma HCC4006 and acute promyelocytic leukemia PL-21, and displayed good pharmacokinetic profiles. Reduction of 3-ketodihydrosphingosine, the direct downstream product of SPT, was confirmed under in vivo settings after oral administration of compounds 4 and 5. Their anti-tumor efficacy was observed in a PL-21 xenograft mouse model. These results suggested that SPT inhibitors might have potential to be effective cancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Serine C-Palmitoyltransferase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , High-Throughput Screening Assays , Humans , Mice , Piperidines/chemical synthesis , Piperidines/chemistry , Piperidines/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Stereoisomerism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
4.
Biochem Biophys Res Commun ; 484(3): 493-500, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28108287

ABSTRACT

Metabolic reprogramming is an essential hallmark of neoplasia. Therefore, targeting cancer metabolism, including lipid synthesis, has attracted much interest in recent years. Serine palmitoyltransferase (SPT) plays a key role in the initial and rate-limiting step of de novo sphingolipid biosynthesis, and inhibiting SPT activity prevents the proliferation of certain cancer cells. Here, we identified a novel and orally available SPT inhibitor, compound-2. Compound-2 showed an anti-proliferative effect in several cancer cell models, reducing the levels of the sphingolipids ceramide and sphingomyelin. In the presence of compound-2, exogenously added S1P partially compensated the intracellular sphingolipid levels through the salvage pathway by partially rescuing compound-2-induced cytotoxicity. This suggested that the mechanism underlying the anti-proliferative effect of compound-2 involved the reduction of sphingolipid levels. Indeed, compound-2 promoted multinuclear formation with reduced endogenous sphingomyelin levels specifically in a compound-2-sensitive cell line, indicating that the effect was induced by sphingolipid reduction. Furthermore, compound-2 showed potent antitumor activity without causing significant body weight loss in the PL-21 acute myeloid leukemia mouse xenograft model. Therefore, SPT may be an attractive therapeutic anti-cancer drug target for which compound-2 may be a promising new drug.


Subject(s)
Antineoplastic Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Serine C-Palmitoyltransferase/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Biological Availability , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , Mouth/metabolism , Treatment Outcome
5.
Am J Respir Cell Mol Biol ; 35(1): 103-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16528013

ABSTRACT

Airway hyperresponsiveness (AHR) is an important feature of bronchial asthma. Although the incidence of AHR has genetic and environmental components, the mechanism of AHR in asthma remains unclear. The identification of genes that are preferentially expressed in a murine model of AHR could help elucidate the molecular mechanisms of this pulmonary pathology. Suppressive subtractive hybridization analysis revealed that eosinophil chemotactic factor by T lymphocytes (ECF-L), a mouse chitinase family protein, was selectively expressed in the lungs of mice with AHR. Induction of ECF-L expression was observed soon after allergen exposure but before the onset of airway inflammation. Cell-specific ECF-L expression was examined by in situ hybridization using digoxigenin-labeled antisense RNA probes and immunofluorescence staining. The assay revealed that the ECF-L-expressing cells in the lungs of the AHR-model mice are alveolar macrophages. Intratracheal administration of an adenoviral vector that expressed antisense ECF-L RNA (Ad-ECF-L-AS) suppressed AHR and eosinophil infiltration. These results indicate that ECF-L may play a critical role in allergic inflammation and bronchial asthma.


Subject(s)
Asthma/metabolism , Bronchial Hyperreactivity/metabolism , Chemokines/metabolism , Chemotactic Factors, Eosinophil/metabolism , Disease Models, Animal , T-Lymphocytes/metabolism , Adenoviridae/genetics , Animals , Asthma/immunology , Bronchial Hyperreactivity/immunology , Chemokines/genetics , Chemotactic Factors, Eosinophil/genetics , Gene Expression Profiling , Lung/cytology , Lung/pathology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred BALB C , RNA, Antisense/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trachea/metabolism
6.
Nat Genet ; 34(4): 440-5, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12858174

ABSTRACT

p53R2, which is regulated by tumor suppressor p53, is a small subunit of ribonucleotide reductase. To determine whether it is involved in DNA repair by supplying deoxyribonucleotides (dNTPs) for resting cells in vivo, we generated a strain of mice lacking Rrm2b (encoding p53R2). These mice developed normally until they were weaned but from then on had growth retardation and early mortality. Pathological examination indicated that multiple organs had failed, and all Rrm2b-null mice died from severe renal failure by the age of 14 weeks. TUNEL staining showed a greater number of apoptotic cells in kidneys of 8-week-old Rrm2b-/- mice relative to wild-type mice. p53 was activated in kidney tissues of Rrm2b-/- mice, leading to transcriptional induction of p53 target genes. Rrm2b-/- mouse embryonic fibroblasts (MEFs) became immortal much earlier than Rrm2b+/+ MEFs. dNTP pools were severely attenuated in Rrm2b-/- MEFs under oxidative stress. Rrm2b deficiency caused higher rates of spontaneous mutation in the kidneys of Rrm2b-/- mice. Our results suggest that p53R2 has a pivotal role in maintaining dNTP levels for repair of DNA in resting cells. Impairment of this pathway may enhance spontaneous mutation frequency and activate p53-dependent apoptotic pathway(s) in vivo, causing severe renal failure, growth retardation and early mortality.


Subject(s)
Cell Cycle Proteins , Deoxyribonucleotides/metabolism , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/metabolism , Ribonucleotide Reductases/deficiency , Ribonucleotide Reductases/genetics , Animals , Apoptosis , DNA Repair , Female , Genes, p53 , Kidney/metabolism , Kidney/pathology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Organ Failure/etiology , Multiple Organ Failure/genetics , Multiple Organ Failure/metabolism , Multiple Organ Failure/pathology , Mutation , Oxidative Stress
7.
Am J Respir Crit Care Med ; 165(8): 1132-6, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11956057

ABSTRACT

Mucus overproduction is a clinical feature of asthma. Ca2+-activated Cl- channel 1 (CaCC1) has been identified as a protein that is expressed in intestinal epithelia and that plays an important role in fluid and electrolyte transport. Recently, its mouse counterpart, gob-5, was identified as a key molecule in the induction of murine asthma through mucus overproduction. To elucidate the relationship of CaCC1 to human asthma, we examined CaCC1 expression using real-time quantitative polymerase chain reaction analysis in bronchial tissues from patients with asthma and normal control subjects. The expression of CaCC1 was significantly upregulated in patients with bronchial asthma compared with control subjects. In situ hybridization and immunohistochemical analysis demonstrated that CaCC1 is located in the bronchial epithelium, especially in mucus-producing goblet cells. In vitro transfection of a CaCC1 expression vector into the human mucoepidermoid cell line, NCI-H292, increased mucus production and induced the MUC5AC gene. These results suggest that CaCC1 plays a direct role in mucus production and differentiation in goblet cells and may contribute to the pathogenesis of asthma through its mucus-inducing activity.


Subject(s)
Asthma/metabolism , Bronchi/metabolism , Calcium/metabolism , Chloride Channels/metabolism , Gene Expression Regulation , Asthma/genetics , Asthma/physiopathology , Bronchi/cytology , Cell Line , Chloride Channels/genetics , Goblet Cells/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Mucin 5AC , Mucins , Mucus/metabolism , Polymerase Chain Reaction , Respiratory Mucosa/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...