Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Biol Chem ; 299(8): 104971, 2023 08.
Article in English | MEDLINE | ID: mdl-37380081

ABSTRACT

The expression of trophoblast cell surface antigen-2 (Trop-2) is enhanced in many tumor tissues and is correlated with increased malignancy and poor survival of patients with cancer. Previously, we demonstrated that the Ser-322 residue of Trop-2 is phosphorylated by protein kinase Cα (PKCα) and PKCδ. Here, we demonstrate that phosphomimetic Trop-2 expressing cells have markedly decreased E-cadherin mRNA and protein levels. Consistently, mRNA and protein of the E-cadherin-repressing transcription factors zinc finger E-Box binding homeobox 1 (ZEB1) were elevated, suggesting transcriptional regulation of E-cadherin expression. The binding of galectin-3 to Trop-2 enhanced the phosphorylation and subsequent cleavage of Trop-2, followed by intracellular signaling by the resultant C-terminal fragment. Binding of ß-catenin/transcription factor 4 (TCF4) along with the C-terminal fragment of Trop-2 to the ZEB1 promoter upregulated ZEB1 expression. Of note, siRNA-mediated knockdown of ß-catenin and TCF4 increased the expression of E-cadherin through ZEB1 downregulation. Knockdown of Trop-2 in MCF-7 cells and DU145 cells resulted in downregulation of ZEB1 and subsequent upregulation of E-cadherin. Furthermore, wild-type and phosphomimetic Trop-2 but not phosphorylation-blocked Trop-2 were detected in the liver and/or lung of some nude mice bearing primary tumors inoculated intraperitoneally or subcutaneously with wild-type or mutated Trop-2 expressing cells, suggesting that Trop-2 phosphorylation, plays an important role in tumor cell mobility in vivo, too. Together with our previous finding of Trop-2 dependent regulation of claudin-7, we suggest that the Trop-2-mediated cascade involves concurrent derangement of both tight and adherence junctions, which may drive metastasis of epithelial tumor cells.


Subject(s)
Galectin 3 , beta Catenin , Animals , Humans , Mice , beta Catenin/genetics , beta Catenin/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression Regulation, Neoplastic , MCF-7 Cells , Mice, Nude , RNA, Messenger/genetics , Trophoblasts/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
2.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34663724

ABSTRACT

Although it is held that proinflammatory changes precede the onset of breast cancer, the underlying mechanisms remain obscure. Here, we demonstrate that FRS2ß, an adaptor protein expressed in a small subset of epithelial cells, triggers the proinflammatory changes that induce stroma in premalignant mammary tissues and is responsible for the disease onset. FRS2ß deficiency in mouse mammary tumor virus (MMTV)-ErbB2 mice markedly attenuated tumorigenesis. Importantly, tumor cells derived from MMTV-ErbB2 mice failed to generate tumors when grafted in the FRS2ß-deficient premalignant tissues. We found that colocalization of FRS2ß and the NEMO subunit of the IκB kinase complex in early endosomes led to activation of nuclear factor-κB (NF-κB), a master regulator of inflammation. Moreover, inhibition of the activities of the NF-κB-induced cytokines, CXC chemokine ligand 12 and insulin-like growth factor 1, abrogated tumorigenesis. Human breast cancer tissues that express higher levels of FRS2ß contain more stroma. The elucidation of the FRS2ß-NF-κB axis uncovers a molecular link between the proinflammatory changes and the disease onset.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Breast Neoplasms/immunology , Carcinogenesis , Cytokines/metabolism , Female , Humans , Inflammation/etiology , Inflammation/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , NF-kappa B/metabolism , Pregnancy , Receptor, ErbB-2/metabolism , Retroviridae Infections , Tumor Microenvironment/immunology , Tumor Virus Infections
3.
J Biol Chem ; 291(46): 24105-24120, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27758869

ABSTRACT

Cancer stem cells (CSCs) represent a small subpopulation of self-renewing oncogenic cells. As in many other stem cells, metabolic reprogramming has been implicated to be a key characteristic of CSCs. However, little is known about how the metabolic features of cancer cells are controlled to orchestrate their CSC-like properties. We recently demonstrated that hyaluronan (HA) overproduction allowed plastic cancer cells to revert to stem cell states. Here, we adopted stable isotope-assisted tracing and mass spectrometry profiling to elucidate the metabolic features of HA-overproducing breast cancer cells. These integrated approaches disclosed an acceleration of metabolic flux in the hexosamine biosynthetic pathway (HBP). A metabolic shift toward glycolysis was also evident by quantitative targeted metabolomics, which was validated by the expression profiles of key glycolytic enzymes. Forced expression of glutamine:fructose-6-phosphate amidotransferase 1 (GFAT1), an HBP rate-limiting enzyme, resembled the results of HA overproduction with regard to HIF-1α accumulation and glycolytic program, whereas GFAT1 inhibition significantly decreased HIF-1α protein level in HA-overproducing cancer cells. Moreover, inhibition of the HBP-HIF-1 axis abrogated HA-driven glycolytic enhancement and reduced the CSC-like subpopulation. Taken together, our results provide compelling evidence that HA production regulates the metabolic and CSC-like properties of breast cancer cells via HBP-coupled HIF-1 signaling.


Subject(s)
Hexosamines/biosynthesis , Hyaluronic Acid/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Female , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , Hexosamines/genetics , Hyaluronic Acid/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Nitrogenous Group Transferases/genetics , Nitrogenous Group Transferases/metabolism
4.
J Neurosci ; 34(18): 6164-76, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24790187

ABSTRACT

Hyaluronan (HA), a large anionic polysaccharide (glycosaminoglycan), is a major constituent of the extracellular matrix of the adult brain. To address its function, we examined the neurophysiology of knock-out mice deficient in hyaluronan synthase (Has) genes. Here we report that these Has mutant mice are prone to epileptic seizures, and that in Has3(-/-) mice, this phenotype is likely derived from a reduction in the size of the brain extracellular space (ECS). Among the three Has knock-out models, namely Has3(-/-), Has1(-/-), and Has2(CKO), the seizures were most prevalent in Has3(-/-) mice, which also showed the greatest HA reduction in the hippocampus. Electrophysiology in Has3(-/-) brain slices demonstrated spontaneous epileptiform activity in CA1 pyramidal neurons, while histological analysis revealed an increase in cell packing in the CA1 stratum pyramidale. Imaging of the diffusion of a fluorescent marker revealed that the transit of molecules through the ECS of this layer was reduced. Quantitative analysis of ECS by the real-time iontophoretic method demonstrated that ECS volume was selectively reduced in the stratum pyramidale by ∼ 40% in Has3(-/-) mice. Finally, osmotic manipulation experiments in brain slices from Has3(-/-) and wild-type mice provided evidence for a causal link between ECS volume and epileptiform activity. Our results provide the first direct evidence for the physiological role of HA in the regulation of ECS volume, and suggest that HA-based preservation of ECS volume may offer a novel avenue for development of antiepileptogenic treatments.


Subject(s)
Brain/pathology , Epilepsy/pathology , Extracellular Space/metabolism , Glucuronosyltransferase/deficiency , Hyaluronic Acid/deficiency , Neurons/physiology , Action Potentials/genetics , Animals , Electric Stimulation , Electroencephalography , Epilepsy/genetics , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Space/genetics , Glucuronosyltransferase/genetics , Hyaluronan Synthases , In Vitro Techniques , Mice , Mice, Knockout , Models, Neurological , Mutation/genetics , Nerve Net/metabolism , Nerve Net/pathology , Neurons/drug effects , Phosphopyruvate Hydratase/metabolism , Quinoxalines/pharmacology
5.
J Biol Chem ; 289(38): 26038-26056, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25077968

ABSTRACT

The cancer stem cell (CSC) model suggests that a small subpopulation of cancer cells possesses the ability to self-renew and give rise to malignant progeny that drive cancer progression. Recent reports have also proposed the existence of certain extra- or intracellular signals that allow cancer progenitors to dynamically revert to a stem cell state. However, the mechanisms underlying cancer cell plasticity and CSC expansion are not entirely clear. Our previous studies using a hyaluronan synthase 2 (Has2) transgenic mouse model demonstrated that hyaluronan overproduction caused rapid development of aggressive breast carcinoma at a high incidence. Thus, we hypothesize that hyaluronan overproduction may accelerate cancer progression by expanding CSC subpopulations during cancer development. Primary cancer cells were established from mammary tumors developed in the transgenic mice and subjected to the Hoechst 33342 dye exclusion assay to sort side population (SP) from non-side population (non-SP) cells. Flow cytometric analysis demonstrated the enrichment of CD44(high)/CD24(low) CSC-like cells in the SP fraction of hyaluronan-overproducing cancer cells. This subpopulation exhibited several characteristics that were similar to CSCs, including cancer-initiating and mammosphere-forming abilities. Excess hyaluronan production drove the epithelial-to-mesenchymal transition process defined as the loss of epithelial phenotypes, up-regulation of transforming growth factor ß (TGF-ß), and induction of the epithelial-to-mesenchymal transition-related transcriptional factors Snail and Twist. Inhibition of TGF-ß-Snail signaling or silencing of Twist expression abrogated the entrance into a stem cell state. Taken together, our findings suggest that hyaluronan overproduction allows plastic cancer cell populations to revert to stem cell states via Twist and the TGF-ß-Snail signaling axis.


Subject(s)
Hyaluronic Acid/biosynthesis , Mammary Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/physiology , Twist-Related Protein 1/metabolism , Animals , Cell Proliferation , Enzyme Induction , Epithelial-Mesenchymal Transition , Female , Gene Expression , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , MAP Kinase Signaling System , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Snail Family Transcription Factors , Tumor Cells, Cultured
6.
Biochem Biophys Res Commun ; 443(2): 666-71, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24333423

ABSTRACT

Hyaluronan synthase (HAS) is a unique membrane-associated glycosyltransferase and its activity is lipid dependent. The dependence however is not well understood, especially in vertebrate systems. Here we investigated the functional association of hyaluronan synthesis in a cholesterol-rich membrane-environment. The culture of human dermal fibroblasts in lipoprotein-depleted medium attenuated the synthesis of hyaluronan. The sequestration of cellular cholesterol by methyl-ß-cyclodextrin also decreased the hyaluronan production of fibroblasts, as well as the HAS activity. To directly evaluate the effects of cholesterol on HAS activity, a recombinant human HAS2 protein with a histidine-tag was expressed as a membrane protein by using a baculovirus system, then successfully solubilized, and isolated by affinity chromatography. When the recombinant HAS2 proteins were reconstituted into liposomes composed of both saturated phosphatidylcholine and cholesterol, this provided a higher enzyme activity as compared with the liposomes formed by phosphatidylcholine alone. Cholesterol regulates HAS2 activity in a biphasic manner, depending on the molar ratio of phosphatidylcholine to cholesterol. Furthermore, the activation profiles of different lipid compositions were determined in the presence or absence of cholesterol. Cholesterol had the opposite effect on the HAS2 activity in liposomes composed of phosphatidylethanolamine or phosphatidylserine. Taken together, the present data suggests a clear functional association between HAS activity and cholesterol-dependent alterations in the physical and chemical properties of cell membranes.


Subject(s)
Cholesterol/metabolism , Fibroblasts/metabolism , Glucuronosyltransferase/metabolism , Hyaluronic Acid/biosynthesis , Lipid Metabolism/physiology , Cells, Cultured , Enzyme Activation , Glucuronosyltransferase/chemistry , Humans , Hyaluronan Synthases
7.
Cell Death Dis ; 15(1): 53, 2024 01 15.
Article in English | MEDLINE | ID: mdl-38225221

ABSTRACT

Chronic metabolic stress paradoxically elicits pro-tumorigenic signals that facilitate cancer stem cell (CSC) development. Therefore, elucidating the metabolic sensing and signaling mechanisms governing cancer cell stemness can provide insights into ameliorating cancer relapse and therapeutic resistance. Here, we provide convincing evidence that chronic metabolic stress triggered by hyaluronan production augments CSC-like traits and chemoresistance by partially impairing nucleotide sugar metabolism, dolichol lipid-linked oligosaccharide (LLO) biosynthesis and N-glycan assembly. Notably, preconditioning with either low-dose tunicamycin or 2-deoxy-D-glucose, which partially interferes with LLO biosynthesis, reproduced the promoting effects of hyaluronan production on CSCs. Multi-omics revealed characteristic changes in N-glycan profiles and Notch signaling activation in cancer cells exposed to mild glycometabolic stress. Restoration of N-glycan assembly with glucosamine and mannose supplementation and Notch signaling blockade attenuated CSC-like properties and further enhanced the therapeutic efficacy of cisplatin. Therefore, our findings uncover a novel mechanism by which tolerable glycometabolic stress boosts cancer cell resilience through altered N-glycosylation and Notch signaling activation.


Subject(s)
Hyaluronic Acid , Resilience, Psychological , Humans , Glycosylation , Hyaluronic Acid/metabolism , Neoplasm Recurrence, Local/metabolism , Polysaccharides/metabolism , Dietary Supplements , Neoplastic Stem Cells/metabolism
8.
Carcinogenesis ; 34(7): 1620-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23455376

ABSTRACT

The aryl hydrocarbon receptor (AhR) plays a suppressive role in cecal carcinogenesis by CUL4B/AhR-mediated ubiquitylation and degradation of ß-catenin, which is activated by xenobiotics and natural ligands. AhR-deficient (AhR(-)(/-)) mice develop cecal tumors with severe inflammation. To elucidate whether the tumors develop autonomously in AhR(-/-) mice due to impaired ß-catenin degradation or in association with accelerated inflammation, we performed two kinds of experiments using germ-free (GF) AhR(-/-) mice and compound mutant mice lacking genes for AhR and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), which plays an essential role in caspase-1 activation in inflammasomes. Both GF AhR(-/-) and AhR(-/-)•ASC(-/-) mice showed considerably reduced tumor development compared with that in AhR(-/-) mice albeit in a 'cancer-prone' state with aberrant ß-catenin accumulation. Blocking of the interleukin (IL)-1ß signaling pathway by treatment with a caspase-1 inhibitor, YVAD, reduced cecal tumorigenesis in AhR(-/-) mice. Signal transducers and activators of transcription 3 (STAT3) activation was detected in the cecal epithelium of the AhR(-/-) mice due to enhanced IL-6 production. An inhibitor of the STAT3 signaling pathway, AG490 suppressed the tumor formation. ASC-mediated inflammation was also found to play a critical role in tumor development in Apc(Min/+) mice, a mouse model of familial adenomatous polyposis. Collectively, these results revealed an important role of the bacteria-triggered or ASC-mediated inflammation signaling pathway in the intestinal tumorigenesis of mice and suggest a possible chemical therapeutic intervention, including AhR ligands and inhibitors of the inflammation pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CARD Signaling Adaptor Proteins/metabolism , Cecal Neoplasms/pathology , Inflammation/pathology , Receptors, Aryl Hydrocarbon/metabolism , Adenomatous Polyposis Coli/immunology , Adenomatous Polyposis Coli/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CARD Signaling Adaptor Proteins/genetics , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cecal Neoplasms/immunology , Cell Line , Enzyme Activation , Female , Germ-Free Life , Inflammasomes/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/immunology , Interleukin-6/immunology , Intestines/immunology , Intestines/microbiology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk , Receptors, Aryl Hydrocarbon/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology , beta Catenin/immunology , beta Catenin/metabolism
9.
J Biol Chem ; 287(30): 25419-33, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22654110

ABSTRACT

The contribution of hyaluronan (HA) to the regulatory network of the hematopoietic microenvironment was studied using knock-out mice of three hyaluronan synthase genes (Has1, Has2, and Has3). The number of hematopoietic progenitors was decreased in bone marrow and increased in extramedullary sites of Prx1-Cre;Has2(flox/flox);Has1(-/-);Has3(-/-) triple knock-out (tKO) mice as compared with wild type (WT) and Has1(-/-);Has3(-/-) double knock-out (dKO) mice. In line with this observation, decreased hematopoietic activity was observed in long term bone marrow cultures (LTBMC) from tKO mice, whereas the formation of the adherent layer and generation of hematopoietic cells in WT and dKO cultures was not different. 4-Methylumbelliferone (4MU) was used to pharmacologically inhibit the production of HA in LTBMC. Treatment with 4MU inhibited HA synthesis, decreased expression of HAS2 and HAS3, and eliminated hematopoiesis in LTBMC, and this effect was alleviated by the addition of exogenous HA. Exogenous HA also augmented the cell motility in LTBMC, which correlated with the HA-stimulated production of chemokines and growth factors. Conditioned media from HA-induced LTBMC enhanced the chemotaxis of hematopoietic stem/progenitor cells (HSPC) in response to SDF-1. Exposure of endothelial cells to 4MU decreased their ability to support HSPC rolling and adhesion. In addition, migration of transplanted HSPC into the marrow of 4MU-pretreated mice was lower than in untreated mice. Collectively, the results suggest that HA depletion reduces the ability of the microenvironment to support HSPC, and confirm a role for HA as a necessary regulatory element in the structure of the hematopoietic microenvironment.


Subject(s)
Bone Marrow/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Hyaluronic Acid/metabolism , Stem Cell Niche/physiology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chemotaxis/drug effects , Chemotaxis/physiology , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Humans , Hyaluronan Synthases , Hyaluronic Acid/genetics , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Mice , Mice, Knockout , Stem Cell Niche/drug effects
10.
Biochim Biophys Acta Gen Subj ; 1867(1): 130250, 2023 01.
Article in English | MEDLINE | ID: mdl-36228878

ABSTRACT

Metabolite sensing, a fundamental biological process, plays a key role in metabolic signaling circuit rewiring. Hexosamine biosynthetic pathway (HBP) is a glucose metabolic pathway essential for the synthesis of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), which senses key nutrients and integrally maintains cellular homeostasis. UDP-GlcNAc dynamically regulates protein N-glycosylation and O-linked-N-acetylglucosamine modification (O-GlcNAcylation). Dysregulated HBP flux leads to abnormal protein glycosylation, and contributes to cancer development and progression by affecting protein function and cellular signaling. Furthermore, O-GlcNAcylation regulates cellular signaling pathways, and its alteration is linked to various cancer characteristics. Additionally, recent findings have suggested a close association between HBP stimulation and cancer stemness; an elevated HBP flux promotes cancer cell conversion to cancer stem cells and enhances chemotherapy resistance via downstream signal activation. In this review, we highlight the prominent roles of HBP in metabolic signaling and summarize the recent advances in HBP and its downstream signaling, relevant to cancer.


Subject(s)
Biological Phenomena , Neoplasms , Humans , Hexosamines/metabolism , Biosynthetic Pathways , Acetylglucosamine/metabolism , Neoplasms/metabolism , Uridine Diphosphate
11.
Biol Pharm Bull ; 35(3): 408-12, 2012.
Article in English | MEDLINE | ID: mdl-22382329

ABSTRACT

Appropriate culture models for tissue mast cells are required to determine how they are involved in regulation of local immune responses. We previously established a culture model for cutaneous mast cells, in which bone marrow-derived immature mast cells were co-cultured with Swiss 3T3 fibroblasts in the presence of stem cell factor. In this study, we focused on the roles of hyaluronan, which is produced by the feeder fibroblasts and forms the extracellular matrix during the co-culture period. Hyaluronan synthesis was found to be mediated by hyaluronan synthase 2 (HAS2) expressed in Swiss 3T3 cells. A decreases in the amount of hyaluronan, which was achieved by retroviral expression of short hairpin RNA for Has2 or by addition of hyaluronidase, significantly enhanced the proliferation of the cultured mast cells without any obvious effects on their maturation. Although we previously demonstrated that CD44 is required for proliferation of cutaneous mast cells, the deficiency of hyaluronan did not affect the proliferation of the cultured mast cells that lack CD44. These findings suggest that the extracellular matrix containing hyaluronan may have a potential to restrict proliferation of cutaneous mast cells in a CD44-independent manner.


Subject(s)
Fibroblasts/metabolism , Glucuronosyltransferase/metabolism , Hyaluronic Acid/metabolism , Mast Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Proliferation , Cells, Cultured , Female , Gene Knockdown Techniques , Glucuronosyltransferase/genetics , Hyaluronan Receptors/genetics , Hyaluronan Synthases , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Swiss 3T3 Cells
12.
Rheumatol Int ; 32(6): 1625-32, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21331573

ABSTRACT

Although rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology, the role of IL-1ß and IL-18 in the pathophysiology of RA has been well established. IL-1ß and IL-18 are generated via cleavage of their pro-forms in the presence of the apoptosis-associated speck-like protein containing a caspase recruit domain (ASC), a known adaptor protein that activates procaspase-1. As such, we investigated the involvement of ASC in the progression of murine collagen-induced arthritis (CIA) and collagen antibody-induced arthritis (CAIA) using ASC-deficient (ASC(-/-)) and wild-type (ASC(+/+)) mice. Analyses were performed by immunohistochemistry for tissues and ELISA for sera. We observed an increase in the expression of ASC, as well as IL-1ß and IL-18, in the joints of CIA DBA mice, which indicated that ASC is involved in disease development. Next, we demonstrated that the infiltration of inflammatory cells and cartilage/bone destruction in CIA knee joints were significantly increased in ASC(+/+) mice compared with ASC(-/-) mice. No such differences were noted in ASC(+/+) and ASC(-/-) CAIA mice. In terms of cytokine expression in knee joints, IL-1ß and IL-18 were depressed in ASC-deficient CIA mice compared with wild-type mice, but were similarly expressed in CAIA joints in both mice groups. Taken together, we can conclude that ASC is involved in the development of CIA and plays a role in the priming phase of the immune response to type II collagen.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Collagen Type II/immunology , Cytoskeletal Proteins/metabolism , Knee Joint/immunology , Knee Joint/metabolism , Animals , Antibodies/blood , Apoptosis Regulatory Proteins , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , CARD Signaling Adaptor Proteins , Cartilage/immunology , Cartilage/metabolism , Cartilage/pathology , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Disease Progression , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Inflammation Mediators/blood , Interleukin-18/blood , Interleukin-1beta/blood , Knee Joint/pathology , Mice , Mice, Inbred DBA , Mice, Knockout
13.
PLoS Pathog ; 5(10): e1000643, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19876387

ABSTRACT

In spite of the importance of hyaluronan in host protection against infectious organisms in the alveolar spaces, its role in mycobacterial infection is unknown. In a previous study, we found that mycobacteria interact with hyaluronan on lung epithelial cells. Here, we have analyzed the role of hyaluronan after mycobacterial infection was established and found that pathogenic mycobacteria can grow by utilizing hyaluronan as a carbon source. Both mouse and human possess 3 kinds of hyaluronan synthases (HAS), designated HAS1, HAS2, and HAS3. Utilizing individual HAS-transfected cells, we show that HAS1 and HAS3 but not HAS2 support growth of mycobacteria. We found that the major hyaluronan synthase expressed in the lung is HAS1, and that its expression was increased after infection with Mycobacterium tuberculosis. Histochemical analysis demonstrated that hyaluronan profoundly accumulated in the granulomatous legion of the lungs in M. tuberculosis-infected mice and rhesus monkeys that died from tuberculosis. We detected hyaluronidase activity in the lysate of mycobacteria and showed that it was critical for hyaluronan-dependent extracellular growth. Finally, we showed that L-Ascorbic acid 6-hexadecanoate, a hyaluronidase inhibitor, suppressed growth of mycobacteria in vivo. Taken together, our data show that pathogenic mycobacteria exploit an intrinsic host-protective molecule, hyaluronan, to grow in the respiratory tract and demonstrate the potential usefulness of hyaluronidase inhibitors against mycobacterial diseases.


Subject(s)
Host-Pathogen Interactions/physiology , Hyaluronic Acid/metabolism , Mycobacterium tuberculosis/physiology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Colony Count, Microbial , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Glycosaminoglycans/pharmacology , Histocytochemistry , Humans , Hyaluronan Synthases , Hyaluronic Acid/pharmacology , Lung/chemistry , Lung/metabolism , Lung/microbiology , Macaca mulatta , Male , Mice , Mycobacterium bovis/physiology , Mycobacterium tuberculosis/metabolism , Rats
14.
Semin Cancer Biol ; 18(4): 268-74, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450474

ABSTRACT

Microenvironmental stimuli can influence the malignant phenotype of cancer cells. Notably, the altered biosynthesis of hyaluronan, a constituent of the extracellular microenvironment, has been implicated in the progress and metastasis of carcinomas. The discovery of hyaluronan synthase (HAS) genes, which encode the key enzymes in hyaluronan biosynthesis, has enabled great strides in understanding the mechanism underlying altered hyaluronan production in cancer and the involvement of this polysaccharide in tumor progression. Recent studies using HAS transgenic mice have provided evidence that overproduction of hyaluronan in mammary tumors accelerates tumor growth through the recruitment of stromal cells and vasculature, revealing further insight into how increased hyaluronan influences the malignant behaviors of cancer cells.


Subject(s)
Hyaluronic Acid/biosynthesis , Neoplasms/metabolism , Animals , Disease Progression , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Stromal Cells/metabolism , Stromal Cells/pathology
15.
Biomolecules ; 10(11)2020 11 07.
Article in English | MEDLINE | ID: mdl-33171800

ABSTRACT

As a major polysaccharide component of the extracellular matrix, hyaluronan plays essential roles in the organization of tissue architecture and the regulation of cellular functions, such as cell proliferation and migration, through interactions with cell-surface receptors and binding molecules. Metabolic pathways for biosynthesis and degradation tightly control the turnover rate, concentration, and molecular size of hyaluronan in tissues. Despite the relatively simple chemical composition of this polysaccharide, its wide range of molecular weights mediate diverse functions that depend on molecular size and tissue concentration. Genetic engineering and pharmacological approaches have demonstrated close associations between hyaluronan metabolism and functions in many physiological and pathological events, including morphogenesis, wound healing, and inflammation. Moreover, emerging evidence has suggested that the accumulation of hyaluronan extracellular matrix and fragments due to the altered expression of hyaluronan synthases and hyaluronidases potentiates cancer development and progression by remodeling the tumor microenvironment. In addition to the well-known functions exerted by extracellular hyaluronan, recent metabolomic approaches have also revealed that its synthesis can regulate cellular functions via the reprogramming of cellular metabolism. This review highlights the current advances in knowledge on the biosynthesis and catabolism of hyaluronan and describes the diverse functions associated with hyaluronan metabolism.


Subject(s)
Hyaluronic Acid/metabolism , Animals , Humans , Hyaluronic Acid/biosynthesis
16.
Lab Invest ; 89(4): 446-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19204665

ABSTRACT

By using the recently established culture system that reproduces the terminal differentiation process of connective tissue-type mast cells, we found significant transcriptional induction of CD44. As CD44 is a primary receptor for hyaluronan (HA), which is one of the major extracellular matrix components, we investigated the role of CD44 in cutaneous mast cells. When co-cultured with fibroblasts, mouse bone marrow-derived cultured mast cells (BMMCs) were found to form clusters in an HA-dependent manner. As compared with BMMCs derived from the wild-type mice, those from the CD44(-/-) mice exhibited impaired growth during the co-cultured period. Furthermore, in the peritoneal cavities and ear tissues, mature mast cells were fewer in number in the CD44(-/-) mice than in the wild-type mice. We investigated roles of CD44 in mast cell proliferation by reconstituting BMMCs into the tissues of mast cell-deficient, Kit(W)/Kit(W-v) mice, and found that the number of metachromatic cells upon acidic toluidine blue staining in the tissues transplanted with CD44(-/-) BMMCs was not significantly changed for 10 weeks, whereas that in the tissues transplanted with the CD44(+/+) BMMCs was significantly increased. These results suggest that CD44 plays a crucial role in the regulation of the cutaneous mast cell number.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Hyaluronan Receptors/physiology , Mast Cells/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Cells, Cultured , Coculture Techniques , Crosses, Genetic , Female , Fibroblasts/cytology , Fibroblasts/physiology , Gene Knock-In Techniques , Hyaluronan Receptors/genetics , Male , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Skin/cytology
17.
J Histochem Cytochem ; 57(4): 327-38, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19064716

ABSTRACT

Apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) is an adaptor molecule activating caspase-1 that stimulates pro-interleukin-1beta (pro-IL-1beta) and pro-IL-18, two pro-inflammatory cytokines with critical functions in host defense against a variety of pathogens. In this study, we investigated the role of ASC in the host defense against Helicobacter pylori utilizing ASC-deficient mice. Mice were orally inoculated with H. pylori; bacterial load, degree of gastritis, and mucosal levels of inflammatory cytokines were analyzed and compared with those obtained from wild-type mice. We found more prominent H. pylori colonization in ASC-deficient mice, as revealed by colony-forming unit counts. Both groups of mice developed gastritis; however, ASC-deficient mice showed significant attenuation of inflammation despite high H. pylori colonization. ELISA, immunohistochemistry, and quantitative RT-PCR analyses revealed complete suppression of IL-1beta and IL-18, and substantial reduction of interferon-gamma (IFN-gamma) expression, in ASC-deficient mice without apparent upregulation of other cytokines, including IL-10 and tumor necrosis factor-alpha. These results as a whole indicate that ASC exerts considerable influence on the host defense, acting through IL-1beta/IL-18 and subsequent IFN-gamma production, which in turn contributes to continuous chronic inflammatory response and consequent reduction of H. pylori colonization.


Subject(s)
Cytoskeletal Proteins/physiology , Helicobacter Infections/immunology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Cytoskeletal Proteins/genetics , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Interferon-gamma/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Knockout
18.
Mediators Inflamm ; 2009: 287387, 2009.
Article in English | MEDLINE | ID: mdl-19759850

ABSTRACT

The apoptosis-associated speck-like protein containing a caspase recruit domain (ASC) is involved in apoptosis and innate immunity and is a major adaptor molecule responsible for procaspase-1 activation. ASC mRNA is encoded by three exons: exons 1 and 3 encode a pyrin domain (PYD) and caspase recruit domain (CARD), respectively, and exon 2 encodes a proline and glycine-rich (PGR) domain. Here, we identified a variant ASC protein (vASC) lacking the PGR domain that was smaller than full length ASC (fASC) derived from fully transcribed mRNA and searched for differences in biochemical and biological nature. Both fASC and vASC were found to activate procaspase-1 to a similar degree, but the efficiency of IL-1beta excretion was significantly higher for vASC. There was also a marked structural difference observed in the fibrous aggregates formed by fASC and vASC. These results suggest that although the PGR domain is dispensable for procaspase-1 activation, it plays an important role in the regulation of the molecular structure and activity of ASC.


Subject(s)
Alternative Splicing , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Interleukin-1beta/metabolism , Blotting, Western , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , HL-60 Cells , Humans , Mass Spectrometry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Biochem ; 165(5): 387-390, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30649348

ABSTRACT

Aberrant glycosylation on tumour cells has been implicated in tumour immune modulation. A recent article published in The Journal of Biochemistry (Sutoh Yoneyama et al., A mechanism for evasion of CTL immunity by altered O-glycosylation of HLA class I, J. Biochem. 2017;161:479-492) showed that bladder cancer cells evaded cytotoxic T lymphocyte-mediated antitumour immunity by a novel mechanism involving the loss of Core 2 structures on human leukocyte antigen Class I O-glycans and subsequent impairment of galectin-glycan lattice formation. The immunosuppressive action of O-glycans on natural killer cell-mediated tumour immunity is also considered an immune evasion system. Furthermore, sialylated O-glycans have been proposed to play a central role in tumour immune escape by modulating the production of immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. Therefore, a better understanding of how alterations in O-glycosylation influence tumour immune evasion will enable the development of novel and more effective therapeutic options for cancer treatment.


Subject(s)
Tumor Escape , Urinary Bladder Neoplasms/immunology , Galectins/metabolism , Glycosylation , Histocompatibility Antigens Class I/immunology , Humans , Polysaccharides/metabolism , T-Lymphocytes, Cytotoxic/immunology
20.
Cell Death Dis ; 10(11): 803, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31645543

ABSTRACT

The hexosamine biosynthetic pathway (HBP) metabolically regulates dynamic cellular events by linking nutrient availability to numerous signaling networks. Significant alterations in the HBP are often associated with cancer pathogenesis. In this study, we investigated the molecular events underlying cancer pathogenesis associated with enhanced HBP flux. Multidimensional analysis of microarray datasets demonstrated up-regulation of genes encoding HBP enzymes in clinical breast cancers and revealed that co-expression of hyaluronan synthase 2 (HAS2) and glutamine:fructose-6-phosphate amidotransferase (GFAT), a rate-limiting enzyme of the HBP, was strongly correlated with a poor prognosis in advanced cancer patients. Consistently with the clinical data, comparative analyses of distinct breast cancer mouse models demonstrated enhancement of the HBP gene expression in primary carcinoma cells, with elevation of Has2 expression and hyaluronan production in aggressive breast cancer cells. The silencing of GFAT reduced CD44high/CD24low cancer stem cell (CSC)-like subpopulations, aldehyde dehydrogenase-positive cell populations, and mammosphere size, which were further diminished by gene targeting of Has2. Has2 gene disruption reduced the in vivo growth of aggressive cancer cells and attenuated pro-tumorigenic Akt/GSK3ß/ß-catenin signaling and cisplatin resistance. Overall protein O-GlcNAcylation was also elevated in association with HBP enhancement in aggressive cancer cells, and the modification exhibited overlapping but distinct roles from the hyaluronan signal in the regulation of CSC-like features. The current data therefore demonstrate that enhanced hexosamine metabolism drives pro-tumorigenic signaling pathways involving hyaluronan and O-GlcNAcylation in aggressive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Hexosamines/metabolism , Hyaluronic Acid/metabolism , Animals , Female , Humans , Mice , Mice, Transgenic , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL