Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 19(9)2018 Sep 09.
Article in English | MEDLINE | ID: mdl-30205617

ABSTRACT

Cancer is initiated by mutations in critical regulatory genes; however, its progression to malignancy is aided by non-neoplastic cells and molecules that create a permissive environment known as the tumor stroma or microenvironment (TME). Interleukin 33 (IL-33) is a dual function cytokine that also acts as a nuclear factor. IL-33 typically resides in the nucleus of the cells where it is expressed. However, upon tissue damage, necrosis, or injury, it is quickly released into extracellular space where it binds to its cognate receptor suppression of tumorigenicity 2 (ST2)L found on the membrane of target cells to potently activate a T Helper 2 (Th2) immune response, thus, it is classified as an alarmin. While its role in immunity and immune-related disorders has been extensively studied, its role in tumorigenesis is only beginning to be elucidated and has revealed opposing roles in tumor development. The IL-33/ST2 axis is emerging as a potent modulator of the TME. By recruiting a cohort of immune cells, it can remodel the TME to promote malignancy or impose tumor regression. Here, we review its multiple functions in various cancers to better understand its potential as a therapeutic target to block tumor progression or as adjuvant therapy to enhance the efficacy of anticancer immunotherapies.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Signal Transduction , Animals , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment
2.
Mol Carcinog ; 56(1): 272-287, 2017 01.
Article in English | MEDLINE | ID: mdl-27120577

ABSTRACT

Liver metastasis is the major cause of death from colorectal cancer (CRC). Understanding its mechanisms is necessary for timely diagnosis and development of effective therapies. Interleukin-33 (IL-33) is an IL-1 cytokine family member that uniquely functions as a cytokine and nuclear factor. It is released by necrotic epithelial cells and activated innate immune cells, functioning as an alarmin or an early danger signal. Its role in invoking type 2 immune response has been established; however, it has contrasting roles in tumor development and metastasis. We identified IL-33 as a potently upregulated cytokine in a highly metastatic murine CRC cell line and examined its role in tumor growth and metastasis to the liver. IL-33 was transgenically expressed in murine and human adenocarcinoma and carcinoma cell lines and their growth and spontaneous metastasis to the liver were assessed in orthotopic models of CRC in wild-type C57Bl/6 and Il33 knockout mice. The results showed that increased expression of IL-33 in CRC cells enhanced their tumor take, growth, and liver metastasis. Tumor- rather than host-derived IL-33 induced the enhanced recruitment of CD11b+ GR1+ and CD11b+ F4/80+ myeloid cells to remodel the tumor microenvironment by increased expression of mobilizing cytokines, and tumor angiogenesis by activating endothelial cells. IL-33 expression was elevated in patient tumor tissues, induced early in adenoma development, and activated by pro-inflammatory cytokines derived from the tumor microenvironment. The data suggest that tumor-derived IL-33 modulates the tumor microenvironment to potently promote colon carcinogenesis and liver metastasis, underscoring its potential as a therapeutic target. © 2016 Wiley Periodicals, Inc.


Subject(s)
Colon/pathology , Colorectal Neoplasms/pathology , Interleukin-33/immunology , Liver Neoplasms/secondary , Liver/pathology , Rectum/pathology , Tumor Microenvironment , Animals , Cell Line, Tumor , Colon/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Humans , Interleukin-33/analysis , Interleukin-33/genetics , Liver/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Rectum/immunology
3.
Immunology ; 149(1): 35-47, 2016 09.
Article in English | MEDLINE | ID: mdl-27238498

ABSTRACT

Microbial infections are a major cause of infant mortality as a result of limitations in immune defences. Interleukin-27 (IL-27) is a heterodimeric cytokine produced primarily by leucocytes and is immunosuppressive toward lymphocytes and leucocytes. Our laboratory demonstrated that human neonatal macrophages express IL-27 more abundantly than adult macrophages. Similarly in mice, IL-27 expression is elevated early in life and maintained through infancy. To determine IL-27-regulated mechanisms that may limit immunity, we evaluated the expression of a number of genes in response to this cytokine in primary human neonatal macrophages. Indoleamine 2,3-dioxygenase (IDO) gene expression was increased dose-responsively by IL-27. We have previously demonstrated inhibition of T-cell proliferation and cytokine production by neonatal macrophage-generated IL-27, and IDO is often implicated in this negative regulation. An increase in IDO protein was demonstrated by immunofluorescence microscopy and was consistent with increased enzyme activity following treatment with IL-27. Inclusion of a soluble receptor to neutralize endogenous IL-27, decreased IDO expression and activity compared with untreated macrophages. In response to IL-27, neonatal macrophages phosphorylate signal transdcuer and activator of transcription 1 (STAT-1) and STAT-3. Both transcription factors are recruited to the IDO regulatory region. STAT-3 dominates during steady-state regulation by lower levels of endogenous IL-27 production. A shift to enhanced STAT-1 recruitment occurs during increased levels of exogenously supplied IL-27. These data suggest an interesting interplay of STAT-1 and STAT-3 to regulate IDO activity and immunosuppression in response to different levels of IL-27 in the microenvironment of the immune response that may further our understanding of this interesting cytokine.


Subject(s)
Interleukin-27/metabolism , Macrophages/immunology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Adult , Animals , Cell Proliferation , Cells, Cultured , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Infant, Newborn , Mice , Mice, Inbred C57BL , Signal Transduction
4.
Mol Pharm ; 11(6): 1897-905, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24779647

ABSTRACT

Ideal "smart" nanoparticles for drug delivery should enhance therapeutic efficacy without introducing side effects. To achieve that, we developed a drug delivery system (HCN) based on a polymer-drug conjugate of poly[2-(pyridin-2-yldisulfanyl)]-graft-poly(ethylene glycol) and camptothecin with an intracellularly cleavable linker and human epidermal growth factor receptor 2 (HER2) targeting ligands. An in vitro drug release study found that HCN was stable in the physiological environment and supersensitive to the stimulus of elevated intracellular redox potential, releasing all payloads in less than 30 min. Furthermore, confocal microscopy revealed that HCN could specifically enter HER2-positive cancer cells. As a consequence, HCN could effectively kill HER2-positive cancer cells while not affecting HER2-negative cells.


Subject(s)
Breast Neoplasms/drug therapy , Camptothecin/administration & dosage , Camptothecin/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Receptor, ErbB-2/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Female , HCT116 Cells , Humans , KB Cells , Oxidation-Reduction , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polymers/administration & dosage , Polymers/chemistry
5.
Clin Exp Metastasis ; 30(7): 903-18, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23748471

ABSTRACT

Colorectal cancer (CRC) is the third most frequent cancer and the third leading cause of cancer deaths in the United States (American Cancer Society, Cancer facts and figures 2012, 20121). The major cause of death is metastasis and frequently, the target organ is the liver. Successful metastasis depends on acquired properties in cancer cells that promote invasion and migration, and on multiple interactions between tumors and host-derived cells in the microenvironment. These processes, however, occur asymptomatically, thus, metastasis remains poorly understood and often diagnosed only at the final stage. To facilitate the elucidation of the mechanisms underlying these processes and to identify the molecular regulators, particularly at the early stages, we developed a mouse model of hepatic metastasis of CRC by cecal implantation of a mouse adenocarcinoma cell line in an immune competent host that reliably recapitulates all steps of tumor growth and metastasis within a defined period. By in vivo selection, we isolated cells of varying metastatic potential. The most highly metastatic CT26-FL3 cells produced liver metastasis as early as 10 days after implantation in 90 % of host mice. These cells expressed elevated levels of genes whose products promote invasion, migration, and mobilization of bone marrow derived cells (BMDCs). Mice bearing tumors from CT26-FL3 had elevated serum levels of OPN, MMP9, S100A8, S100A9, SAA3, and VEGFA that promote invasion and BMDC mobilization, and showed enhanced BMDC recruitment to the liver where they established a pre-metastatic niche. This model provides an important platform to characterize metastatic cells and elucidate tumor-host interactions and mechanisms that drive liver metastasis of CRC.


Subject(s)
Colorectal Neoplasms/pathology , Liver Neoplasms, Experimental/secondary , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction
6.
Immunology ; 139(4): 484-93, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23464355

ABSTRACT

Microbial infections are a major cause of infant mortality worldwide because of impaired immune defences in this population. The nature of this work was to further understand the mechanistic limitations of the neonatal and infant immune response. Interleukin-27 (IL-27) is a heterodimeric cytokine of the IL-12 family that is produced primarily by antigen-presenting cells and is immunosuppressive toward a variety of immune cell types. We show that IL-27 gene expression is elevated in cord blood-derived macrophages relative to macrophages originating from healthy adults. We also evaluated the duration over which elevated IL-27 gene expression may impact immune responses in mice. Age-dependent analysis of IL-27 gene expression indicated that levels of IL-27 remained significantly elevated throughout infancy and then declined in adult mice. Flow cytometric analysis of intracellular cytokine-stained splenocytes further confirmed these results. Interleukin-27 may be induced during pregnancy to contribute to the immunosuppressive environment at the fetal-maternal interface because we demonstrate dose-responsive gene expression to progesterone in macrophages. Neutralization of IL-27 in neonatal macrophages improved the ability of these cells to limit bacterial replication. Moreover, neutralization of IL-27 during incubation with the Mycobacterium bovis bacillus Calmette-Guérin vaccine augmented the level of interferon-γ elicited from allogeneic CD4+ T lymphocytes. This suggests that blocking IL-27 during vaccination and infection may improve immune responses in newborn and infant populations. Furthermore, mice will be a suitable model system to further address these possibilities.


Subject(s)
Fetal Blood/immunology , Immune Tolerance , Interleukins/metabolism , Macrophages/immunology , Age Factors , Aging/immunology , Animals , Animals, Newborn , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Dose-Response Relationship, Drug , Fetal Blood/cytology , Flow Cytometry , Gene Expression Regulation, Developmental , Humans , Immune Tolerance/drug effects , Infant, Newborn , Interferon-gamma/metabolism , Interleukins/genetics , Lymphocyte Activation , Macrophages/drug effects , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium bovis/immunology , Progesterone/pharmacology , Spleen/immunology , Time Factors , Up-Regulation
7.
Microsc Microanal ; 17(4): 528-39, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21473808

ABSTRACT

Tumors consist of a heterogeneous population of neoplastic cells infiltrated by an equally heterogeneous collection of nonneoplastic cells that comprise the tumor microenvironment. Tumor growth, invasion, and metastasis depend on multiple interactions between these cells. To assess their potential as therapeutic targets or vehicles for tumor specific delivery of therapeutic agents, we examined the contribution of bone marrow derived cells (BMDCs) to the intestinal tumor microenvironment. Hematopoietic stem cells expressing the enhanced green fluorescent protein (eGFP) were transplanted into lethally irradiated ApcMin/+ mice, and their engraftment was analyzed by confocal microscopy. The results showed abundant infiltration of eGFP cells into the small intestine, colon, and spleen compared to heart, muscle, liver, lung, and kidney. Within the intestine, there was a pronounced gradient of engraftment along the anterior to posterior axis, with enhanced infiltration into adenomas. Immunofluorescence analysis showed that osteopontin was expressed in tumor stromal cells but not in nontumor stromal populations, suggesting that gene expression in these cells is distinct. Tumor vasculature in ApcMin/+ mice was chaotic compared to normal intestinal regions. Our data suggest that BMDCs can be harnessed for tumor-targeted therapies to enhance antitumor efficacy.


Subject(s)
Adenoma/pathology , Hematopoietic System/cytology , Intestinal Neoplasms/pathology , Intestines/cytology , Tumor Microenvironment , Animals , Bone Marrow Transplantation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Osteopontin/biosynthesis , Staining and Labeling/methods
8.
BMC Cancer ; 10: 426, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20707908

ABSTRACT

BACKGROUND: We are developing a cross-species comparison strategy to distinguish between cancer driver- and passenger gene alteration candidates, by utilizing the difference in genomic location of orthologous genes between the human and other mammals. As an initial test of this strategy, we conducted a pilot study with human colorectal cancer (CRC) and its mouse model C57BL/6J ApcMin/+, focusing on human 5q22.2 and 18q21.1-q21.2. METHODS: We first performed bioinformatics analysis on the evolution of 5q22.2 and 18q21.1-q21.2 regions. Then, we performed exon-targeted sequencing, real time quantitative polymerase chain reaction (qPCR), and real time quantitative reverse transcriptase PCR (qRT-PCR) analyses on a number of genes of both regions with both human and mouse colon tumors. RESULTS: These two regions (5q22.2 and 18q21.1-q21.2) are frequently deleted in human CRCs and encode genuine colorectal tumor suppressors APC and SMAD4. They also encode genes such as MCC (mutated in colorectal cancer) with their role in CRC etiology unknown. We have discovered that both regions are evolutionarily unstable, resulting in genes that are clustered in each human region being found scattered at several distinct loci in the genome of many other species. For instance, APC and MCC are within 200 kb apart in human 5q22.2 but are 10 Mb apart in the mouse genome. Importantly, our analyses revealed that, while known CRC driver genes APC and SMAD4 were disrupted in both human colorectal tumors and tumors from ApcMin/+ mice, the questionable MCC gene was disrupted in human tumors but appeared to be intact in mouse tumors. CONCLUSIONS: These results indicate that MCC may not actually play any causative role in early colorectal tumorigenesis. We also hypothesize that its disruption in human CRCs is likely a mere result of its close proximity to APC in the human genome. Expanding this pilot study to the entire genome may identify more questionable genes like MCC, facilitating the discovery of new CRC driver gene candidates.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, APC/physiology , Genes, MCC/physiology , Genome, Human , Animals , Base Sequence , Chromosomes, Human, Pair 18/genetics , Chromosomes, Human, Pair 5/genetics , Computational Biology , Evolution, Molecular , Genes, DCC/physiology , Humans , Mice , Molecular Sequence Data , Pilot Projects , Smad4 Protein/genetics , Species Specificity
9.
J Appl Physiol (1985) ; 109(4): 1155-61, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651218

ABSTRACT

Criteria for diagnosing cachexia in adults include unintentional loss in body weight, decreased strength, fatigue, anorexia, and low muscle mass. Cachexia is also associated with systemic inflammation, altered metabolism, and anemia. The Apc(Min/+) mouse is a model of cachexia directly related to intestinal tumor burden and subsequent chronic inflammation. These mice also demonstrate muscle weakness, fatigue, decreased volitional activity, and elevated circulating IL-6 levels. The purpose of this study was to determine the time course of changes in physical activity and their relationship to anemia, muscle apoptosis, and muscle mass and body mass loss during cachexia. A subset of male Apc(Min/+) mice were given access to voluntary activity wheels from 5 to 26 wk of age, while sedentary male Apc(Min/+) mice were housed in cages lacking wheels. At the study's end mice were stratified by cachectic symptoms. Severely cachectic mice had decreased wheel running performance at 15 wk of age, while anemia and body weight loss were not present until 18 wk of age. Severely cachectic mice had lower hemoglobin levels compared with mildly cachectic mice at 13, 18, and 22 wk of age. Severely cachectic mice also demonstrated threefold more BCL2-associated X protein (BAX) protein in the gastrocnemius muscle at 26 wk of age compared with mildly cachectic mice. In sedentary Apc(Min/+) mice at 26 wk of age anemia was present, and markers of apoptosis were induced in severely cachectic muscle. Proapoptotic protein expression was induced in both red and white portions of gastrocnemius muscle as well as in soleus muscle of severely cachectic mice compared with mildly cachectic mice. These data demonstrate that decrements in wheel running performance precede loss of body mass and that inherent muscle oxidative capacity is not protective against muscle apoptosis.


Subject(s)
Adenomatous Polyposis Coli/genetics , Apoptosis , Cachexia/genetics , Genes, APC , Motor Activity , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Mutation , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli/physiopathology , Anemia/blood , Anemia/genetics , Animals , Cachexia/metabolism , Cachexia/pathology , Cachexia/physiopathology , Disease Models, Animal , Gene Expression Regulation , Hemoglobins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Skeletal/metabolism , Oxidation-Reduction , Phenotype , Running , Severity of Illness Index , Time Factors , Weight Loss , bcl-2-Associated X Protein/metabolism
10.
Biochemistry ; 49(11): 2475-82, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20151707

ABSTRACT

Human and other mammalian thymidylate synthase (TS) enzymes have an N-terminal extension of approximately 27 amino acids that is not present in bacterial TSs. The extension, which is disordered in all reported crystal structures of TSs, has been considered to play a primary role in protein turnover but not in catalytic activity. In mammalian cells, the variant V3A has a half-life similar to that of wild-type human TS (wt hTS) while V3T is much more stable; V3L, V3F, and V3Y have half-lives approximately half of that for wt hTS. Catalytic turnover rates for most Val3 mutants are only slightly diminished, as expected. However, two mutants, V3L and V3F, have strongly compromised dUMP binding, with K(m,app) values increased by factors of 47 and 58, respectively. For V3L, this observation can be explained by stabilization of the inactive conformation of the loop of residues 181-197, which prevents substrate binding. In the crystal structure of V3L, electron density corresponding to a leucine residue is present in a position that stabilizes the loop of residues 181-197 in the inactive conformation. Since this density is not observed in other mutants and all other leucine residues are ordered in this structure, it is likely that this density represents Leu3. In the crystal structure of a V3F.FdUMP binary complex, the nucleotide is bound in an alternative mode to that proposed for the catalytic complex, indicating that the high K(m,app) value is caused not by stabilization of the inactive conformer but by substrate binding in a nonproductive, inhibitory site. These observations show that the N-terminal extension affects the conformational state of the hTS catalytic region. Each of the mechanisms leading to the high K(m,app) values can be exploited to facilitate design of compounds acting as allosteric inhibitors of hTS.


Subject(s)
Amino Acid Substitution , Intracellular Space/enzymology , Thymidylate Synthase/chemistry , Thymidylate Synthase/metabolism , Valine , Animals , Cell Line , Cricetinae , Crystallography, X-Ray , Enzyme Stability , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Thymidylate Synthase/genetics , Transformation, Bacterial
11.
J Biol Chem ; 284(46): 31597-607, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19797058

ABSTRACT

The ubiquitin-independent proteasomal degradation pathway is increasingly being recognized as important in regulation of protein turnover in eukaryotic cells. One substrate of this pathway is the pyrimidine biosynthetic enzyme thymidylate synthase (TS; EC 2.1.1.45), which catalyzes the reductive methylation of dUMP to form dTMP and is essential for DNA replication during cell growth and proliferation. Previous work from our laboratory showed that degradation of TS is ubiquitin-independent and mediated by an intrinsically disordered 27-residue region at the N-terminal end of the molecule. In the current study we show that this region, in cooperation with an alpha-helix formed by the next 15 residues, functions as a degron, i.e. it is capable of destabilizing a heterologous protein to which it is fused. Comparative analysis of the primary sequence of TS from a number of mammalian species revealed that the N-terminal domain is hypervariable among species yet is conserved with regard to its disordered nature, its high Pro content, and the occurrence of Pro at the penultimate site. Characterization of mutant proteins showed that Pro-2 protects the N terminus against N(alpha)-acetylation, a post-translational process that inhibits TS degradation. However, although a free amino group at the N terminus is necessary, it is not sufficient for degradation of the polypeptide. The implications of these findings to the proteasome-targeting function of the N-terminal domain, particularly with regard to its intrinsic flexibility, are discussed.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Thymidylate Synthase/chemistry , Thymidylate Synthase/metabolism , Ubiquitin/metabolism , Acetylation , Amino Acid Sequence , Animals , Cells, Cultured , Cricetinae , Cricetulus , Electrophoresis, Gel, Two-Dimensional , Fibroblasts/enzymology , Humans , Immunoblotting , Lung/cytology , Lung/enzymology , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thymidylate Synthase/genetics
12.
Cancer Prev Res (Phila) ; 2(7): 641-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19549797

ABSTRACT

Diet and exercise are two environmental factors that can alter colon cancer risk. The purpose of this study was to determine if regular moderate-intensity treadmill exercise training could attenuate polyp formation in Apc(Min/+) mice fed the Western-style diet. Four-week-old male Apc(Min/+) mice (n = 12 per group) were assigned to AIN-76A Control, AIN-76A Exercise, Western Control, or Western Exercise treatment groups. Mice were weaned to these diets and either subjected to regular moderate-intensity treadmill exercise (18 m/min, 60 min/d, 6 d/wk) or remained sedentary for 6 weeks. Mice fed the Western-style diet consumed approximately 14% more calories and had 42% more epididymal fat compared with mice fed the AIN-76A diet. Exercise had no effect on fat pad mass with either diet treatment. Exercise reduced total intestinal polyp number by 50% and the number of large polyps (>1 mm diameter) by 67% in AIN-76A-fed mice. The Western-style diet increased polyp number by 75% when compared with AIN-76A-fed mice, but exercise did not decrease polyp number or alter polyp size in mice fed the Western-style diet. Markers of systemic inflammation and immune system function were improved with exercise in mice fed the AIN-76A diet. Mice fed the Western-style diet showed more inflammation and immunosuppression, which were not completely ameliorated by exercise. These data suggest that the induction of adiposity, inflammation, and immunosuppression by the Western-style diet may compromise the beneficial effect of moderate-intensity exercise on the intestinal polyp burden in Apc(Min/+) mice.


Subject(s)
Colonic Neoplasms/genetics , Intestinal Polyps/pathology , Animals , Body Weight , Colorectal Neoplasms , Diet , Female , Immunosuppression Therapy , Inflammation , Intestinal Polyps/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Physical Conditioning, Animal , Polyps
13.
J Biochem ; 145(3): 289-97, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19106159

ABSTRACT

Proliferation of intestinal epithelial cells is rhythmic throughout the day. This temporal organization occurs through the interaction between the endogenous peripheral circadian clock and pathways controlling cell cycle progression. Per2, a core clock gene with tumour suppresser function, is critical to clock function and to the regulation of cellular proliferation. Circadian disruption, which increases colon cancer incidence, may do so by deregulating clock controlled epithelial cell proliferation. Increased expression of beta-catenin is a contributing cause of most familial and spontaneous human colon cancer and the cause of multiple intestinal neoplasia of the Apc(Min/+) mouse. Here we report that increased beta-catenin destabilizes PER2 clock protein by inducing beta-TrCP, an F-box protein of SCF ubiquitin E3 ligase. In the intestinal mucosa of the Apc(Min/)(+) mouse, the decrease in PER2 protein levels is associated with altered circadian rhythms of clock genes, Per1 and Per2, and clock controlled genes, Dbp and Wee1. These findings suggest that disruption of the peripheral intestinal circadian clock may be intimately involved in beta-catenin induced intestinal epithelial neoplastic transformation in both mouse and man.


Subject(s)
Cell Cycle Proteins/metabolism , Circadian Rhythm/genetics , Intestinal Mucosa/metabolism , Nuclear Proteins/metabolism , Trans-Activators/genetics , Transcription Factors/metabolism , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Adenomatous Polyposis Coli/metabolism , Animals , CLOCK Proteins , Down-Regulation , HCT116 Cells , Humans , Mice , Mice, Inbred C57BL , Motor Activity , Period Circadian Proteins , Protein Processing, Post-Translational , Protein Stability
14.
Pflugers Arch ; 457(5): 989-1001, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18712412

ABSTRACT

Interleukin-6 (IL-6) is necessary for cachexia in Apc ( Min/+ ) mice, but the mechanisms inducing this myofiber wasting have not been established. The purpose of this study was to examine gastrocnemius muscle wasting in the Apc ( Min/+ ) mouse and to determine IL-6 regulated mechanisms contributing to muscle loss. Gastrocnemius type IIB mean fiber cross-sectional area (CSA) from Apc ( Min/+ ) mice decreased 32% between 13 and 22 weeks of age. Apc ( Min/+ ) mice lacking IL-6 did not have type IIB fiber atrophy, while overexpression of circulating IL-6 exacerbated the loss of type IIB fiber CSA in Apc ( Min/+ ) mice. Muscle Atrogin-I mRNA expression was induced at least ninefold at 18 and 22 weeks of age compared to 13-week-old mice. Atrogin-I gene expression was also induced by overexpression of circulating IL-6. These data suggest that high circulating IL-6 levels induce type IIB fiber CSA loss in Apc ( Min/+ ) mice, and circulating IL-6 is sufficient to regulate Atrogin-I gene expression in cachectic mice.


Subject(s)
Cachexia/physiopathology , Interleukin-6/physiology , Muscle Proteins/biosynthesis , Muscular Atrophy/physiopathology , SKP Cullin F-Box Protein Ligases/biosynthesis , Animals , Cachexia/pathology , Gene Expression , Interleukin-6/blood , Intestinal Polyps/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Muscle Proteins/metabolism , Muscle Strength/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/pathology , Myofibrils/pathology
15.
Mol Cancer Res ; 6(11): 1786-93, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19010825

ABSTRACT

Colorectal cancer risk is increased in shift workers with presumed circadian disruption. Intestinal epithelial cell proliferation is gated throughout each day by the circadian clock. Period 2 (Per2) is a key circadian clock gene. Per2 mutant (Per2(m/m)) mice show an increase in lymphomas and deregulated expression of cyclin D and c-Myc genes that are key to proliferation control. We asked whether Per2 clock gene inactivation would accelerate intestinal and colonic tumorigenesis. The effects of PER2 on cell proliferation and beta-catenin were studied in colon cancer cell lines by its down-regulation following RNA interference. The effects of Per2 inactivation in vivo on beta-catenin and on intestinal and colonic polyp formation were studied in mice with Per2 mutation alone and in combination with an Apc mutation using polyp-prone Apc(Min/+) mice. Down-regulation of PER2 in colon cell lines (HCT116 and SW480) increases beta-catenin, cyclin D, and cell proliferation. Down-regulation of beta-catenin along with Per2 blocks the increase in cyclin D and cell proliferation. Per2(m/m) mice develop colonic polyps and show an increase in small intestinal mucosa beta-catenin and cyclin D protein levels compared with wild-type mice. Apc(Min/+)Per2(m/m) mice develop twice the number of small intestinal and colonic polyps, with more severe anemia and splenomegaly, compared with Apc(Min/+) mice. These data suggest that Per2 gene product suppresses tumorigenesis in the small intestine and colon by down-regulation of beta-catenin and beta-catenin target genes, and this circadian core clock gene may represent a novel target for colorectal cancer prevention and control.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Colonic Neoplasms/pathology , Genes, APC , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Circadian Rhythm , Colon/pathology , Colonic Neoplasms/genetics , Colonic Polyps/pathology , Cyclin D , Cyclins/metabolism , Disease Models, Animal , Down-Regulation , Humans , Intestinal Mucosa/pathology , Intestinal Polyps/pathology , Mice , Neoplasm Invasiveness/genetics , Period Circadian Proteins , RNA Interference , beta Catenin/genetics
16.
J Appl Physiol (1985) ; 104(4): 1137-43, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18239078

ABSTRACT

Many epidemiological studies have demonstrated that level of exercise is associated with reduced colorectal cancer risk. Treadmill training can decrease Apc(Min/+) mouse intestinal polyp number and size, but the mechanisms remain unclear. Understanding the molecular changes in the tumor following exercise training may provide insight on the mechanism by which exercise decreases Apc(Min/+) mouse polyp formation and growth. The purpose of this study was to determine if exercise can modulate Apc(Min/+) mouse intestinal polyp cellular signaling related to tumor formation and growth. Male Apc(Min/+) mice were randomly assigned to control (n = 20) or exercise (n = 20) treatment groups. Exercised mice ran on a treadmill at a moderate intensity (18 m/min, 60 min, 6 days/wk, 5% grade) for 9 wk. Polyps from Apc(Min/+) mice were used to quantify markers of polyp inflammation, apoptosis, and beta-catenin signaling. Exercise decreased the number of macrophages in polyps by 35%. Related to apoptosis, exercise decreased the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells by 73% in all polyps. Bax protein expression in polyps was decreased 43% by exercise. beta-Catenin phosphorylation was elevated 3.3-fold in polyps from exercised mice. Moderate-intensity exercise training alters cellular pathways in Apc(Min/+) mouse polyps, and these changes may be related to the exercise-induced reduction in polyp formation and growth.


Subject(s)
Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Physical Conditioning, Animal/physiology , Animals , Apoptosis/physiology , Blotting, Western , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Inflammation/pathology , Inflammation/physiopathology , Macrophages/physiology , Male , Mice , Mice, Knockout , Neutrophil Infiltration/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
17.
Am J Physiol Regul Integr Comp Physiol ; 294(2): R393-401, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18056981

ABSTRACT

The Apc(Min/+) mouse has a mutation in the Apc tumor suppressor gene and develops intestinal polyps, beginning at 4 wk of age. This mouse develops cachexia by 6 mo, characterized by significant loss of muscle and fat tissue. The purpose of the present study was to determine the role of circulating interleukin-6 (IL-6) and the polyp burden for the development of cachexia in Apc(Min/+) mice. At 26 wk of age, mice exhibiting severe cachectic symptoms had a 61% decrease in gastrocnemius muscle weight, complete loss of epididymal fat, a 10-fold increase in circulating IL-6 levels, and an 89% increase in intestinal polyps compared with mildly cachectic animals. Apc(Min/+)/IL-6(-/-) mice did not lose gastrocnemius muscle mass or epididymal fat pad mass while overall polyp number decreased by 32% compared with Apc(Min/+) mice. Plasmid-based IL-6 overexpression in Apc(Min/+)/IL-6(-/-) mice led to a decrease in gastrocnemius muscle mass and epididymal fat pad mass and increased intestinal polyp burden. IL-6 overexpression did not induce cachexia in non-tumor-bearing mice. These data demonstrate that IL-6 is necessary for the onset of adipose and skeletal muscle wasting in the Apc(Min/+) mouse and that circulating IL-6 can regulate Apc(Min/+) mouse tumor burden.


Subject(s)
Adenomatous Polyposis Coli/immunology , Cachexia/immunology , Genes, APC , Interleukin-6/immunology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adipose Tissue/pathology , Animals , Body Weight , Cachexia/genetics , Cachexia/pathology , Electroporation , Gene Expression/immunology , Interleukin-6/blood , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Motor Activity , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Plasmids , STAT3 Transcription Factor/metabolism , Severity of Illness Index
18.
Int J Exp Pathol ; 87(3): 227-36, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16709231

ABSTRACT

Germ-line mutation of the Apc gene has been linked to familial adenomatous polyposis (FAP) that predisposes to colon cancer. Apc(Min/+) mice, heterozygous for the Apc gene mutation, progressively develop small intestinal tumours in a manner that is analogous to that observed in the colon of patients with FAP (Su et al. 1992; Fodde et al. 1994; Moser et al. 1995). We have studied the effects of Apc gene mutation on murine intestinal and extra-intestinal, proliferatively active tissues. We have contrasted the histology to that of the age- and sex-matched wild-type C57BL/6 mice. Histological assessment of the normal appearing intestinal mucosa demonstrates minimal change in size of crypts. In contrast, villi are longer in the ileum of Apc(Min/+) mice relative to C57BL/6 mice at 12 and 15 weeks of age. Vigorous splenic haematopoiesis in Apc(Min/+) mice was seen at 12 and 15 weeks of age, as reflected by marked splenomegaly, increased splenic haematopoietic cells and megakaryocytes. Peripheral blood counts, however, did not differ between C57BL/6 and Apc(Min/+) mice at 15 weeks of age. Lymphoid depletion in Apc(Min/+) mice was characterized by diminished numbers of splenic lymphoid follicles and small intestinal Peyer's patches. The ovaries of 12- and 15-week-old Apc(Min/+) mice exhibited increased numbers of atretic follicles, and estrous cycling by serial vaginal smears showed tendency of elongation in the mutant mice during these age ranges. The testicles of 10-week-old Apc(Min/+) mice showed increased numbers of underdeveloped seminiferous tubules. Collectively, these data suggest that, in addition to its obvious effects upon intestinal adenoma formation, Apc gene mutation causes impairment of developmental and apparent differentiation blockade in proliferative tissues, including those of the haematopoietic system, lymphoid and reproductive tract.


Subject(s)
Adenomatous Polyposis Coli/pathology , Hematopoiesis , Intestinal Mucosa/pathology , Intestines/pathology , Animals , Female , Genes, APC , Germ-Line Mutation , Lymphocyte Count , Lymphocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Animal , Ovary/pathology , Testis/pathology
19.
Biochem J ; 394(Pt 1): 355-63, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16259621

ABSTRACT

Thymidylate synthase (TS) catalyses the reductive methylation of dUMP to form dTMP, a reaction that is essential for maintenance of nucleotide pools during cell growth. Because the enzyme is indispensable for DNA replication in actively dividing cells, it is an important target for cytotoxic drugs used in cancer chemotherapy, including fluoropyrimidines (e.g. 5-fluorouracil and 5-fluoro-2'-deoxyuridine) and anti-folates (e.g. raltitrexed, LY231514, ZD9331 and BW1843U89). These drugs generate metabolites that bind to the enzyme's active site and inhibit catalytic activity, leading to thymidylate deprivation and cellular apoptosis. Ligand binding to TS results in stabilization of the enzyme and an increase in its intracellular concentration. Previously, we showed that degradation of the TS polypeptide is carried out by the 26 S proteasome in a ubiquitin-independent manner. Such degradation is directed by the disordered N-terminal region of the TS polypeptide, and is abrogated by ligand binding. In the present study, we have verified the ubiquitin-independent nature of TS proteolysis by showing that a 'lysine-less' polypeptide, in which all lysine residues were replaced by arginine, is still subject to proteasome-mediated degradation. In addition, we have mapped the structural determinants of intracellular TS degradation in more detail and show that residues at the N-terminal end of the molecule, particularly the penultimate amino acid Pro2, play an important role in governing the half-life of the enzyme. This region is capable on its own of destabilizing an evolutionarily distinct TS molecule that normally lacks this domain, indicating that it functions as a degradation signal. Interestingly, degradation of an intrinsically unstable mutant form of TS, containing a Pro-->Leu substitution at residue 303, is directed by C-terminal, rather than N-terminal, sequences. The implications of these findings for the control of TS expression, and for the regulation of protein degradation in general, are discussed.


Subject(s)
Protein Processing, Post-Translational , Thymidylate Synthase/chemistry , Thymidylate Synthase/metabolism , Amino Acid Sequence , Animals , Cricetinae , Cricetulus , Humans , Lysine , Mutagenesis, Site-Directed , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Signal Transduction , Thymidylate Synthase/genetics , Ubiquitin/physiology
20.
Biochemistry ; 43(7): 1972-9, 2004 Feb 24.
Article in English | MEDLINE | ID: mdl-14967037

ABSTRACT

Thymidylate synthase (EC 2.1.1.45) (TS) catalyzes the conversion of dUMP to dTMP and is therefore indispensable for DNA replication in actively dividing cells. The enzyme is a critical target at which chemotherapeutic agents such as fluoropyrimidines (e.g., 5-fluorouracil and 5-fluoro-2'-deoxyuridine) and folic acid analogues (e.g., raltitrexed, LY231514, ZD9331, and BW1843U89) are directed. These agents exert their effects through the generation of metabolites that bind the active site of TS and inhibit catalytic activity. The binding of ligands to the TS molecule leads to dramatic changes in the conformation of the enzyme, particularly within the C-terminal domain. Stabilization of the enzyme and an increase in its intracellular level are associated with ligand binding and may be important in cellular response to TS-directed drugs. In the present study, we have examined molecular features of the TS molecule that control its degradation. We find that the C-terminal conformational shift is not required for ligand-mediated stabilization of the enzyme. In addition, we demonstrate that the N-terminus of the TS polypeptide, which is extended in the mammalian enzyme and is disordered in crystal structures, is a primary determinant of the enzyme's half-life. Finally, we show that TS turnover is carried out by the 26S proteasome in a ubiquitin-independent manner. These findings provide the basis for a mechanistic understanding of TS degradation and its regulation by antimetabolites.


Subject(s)
Intracellular Fluid/enzymology , Proteasome Endopeptidase Complex , Thymidylate Synthase/chemistry , Thymidylate Synthase/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line , Cricetinae , Enzyme Induction/genetics , Enzyme Stability/genetics , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Hydrolases/chemistry , Protein Conformation , Protein Structure, Tertiary/genetics , Signal Transduction/genetics , Thymidylate Synthase/biosynthesis , Thymidylate Synthase/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...