Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Elife ; 102021 08 09.
Article in English | MEDLINE | ID: mdl-34369876

ABSTRACT

Despite the mechanisms of central and peripheral tolerance, the mature B cell compartment contains cells reactive for self-antigen. How these cells are poised not to respond and the mechanisms that restrain B cell responses to low-affinity endogenous antigens are not fully understood. Here, we demonstrate a critical role for the glycan-binding protein galectin-9 in setting the threshold of B cell activation and that loss of this regulatory network is sufficient to drive spontaneous autoimmunity. We further demonstrate a critical role for galectin-9 in restraining not only conventional B-2 B cells, but also innate-like B-1a cells. We show that galectin-9-deficient mice have an expanded population of B-1a cells and increased titers of B-1a-derived autoantibodies. Mechanistically, we demonstrate that galectin-9 regulates BCR and distinct TLR responses in B-1a cells, but not B-1b cells, by regulating the interaction between BCR and TLRs with the regulatory molecules CD5 and CD180, respectively. In the absence of galectin-9, B-1a cells are more readily activated and secrete increased titers of autoantibodies that facilitate autoantigen delivery to the spleen, driving autoimmune responses.


Subject(s)
Autoimmunity/genetics , B-Lymphocytes/metabolism , Galectins/deficiency , Age Factors , Animals , Germinal Center/metabolism , Germinal Center/pathology , Mice , Nephritis/genetics , Splenomegaly/genetics
2.
J Immunol ; 206(11): 2692-2699, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33963043

ABSTRACT

NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome has been implicated in a variety of inflammatory disorders, and its activation should be tightly controlled to avoid detrimental effects. NLRP3 protein expression is considered as the rate-limiting step for NLRP3 inflammasome activation. In this study, we show that galectin-9 (encoded by lgals9) attenuated NLRP3 inflammasome activation by promoting the protein degradation of NLRP3 in primary peritoneal macrophages of C57BL/6J mice. Lgals9 deficiency enhances NLRP3 inflammasome activation and promotes NLRP3-dependent inflammation in C57BL/6J mice in vivo. Mechanistically, galectin-9 interacts with NLRP3, promotes the formation of NLRP3/p62 (an autophagic cargo receptor, also known as SQSTM1) complex, and thus facilitates p62-dependent autophagic degradation of NLRP3 in primary peritoneal macrophages of C57BL/6J mice and HEK293T cells. Therefore, we identify galectin-9 as an "eat-me" signal for selective autophagy of NLRP3 and uncover the potential roles of galectins in controlling host protein degradation. Furthermore, our work suggests galectin-9 as a priming therapeutic target for the diseases caused by improper NLRP3 inflammasome activation.


Subject(s)
Autophagy/immunology , Galectins/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Galectins/deficiency , Galectins/genetics , HEK293 Cells , Humans , Inflammasomes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , THP-1 Cells
3.
Sci Rep ; 11(1): 5991, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727589

ABSTRACT

The adipose tissue is regarded as an endocrine organ and secretes bioactive adipokines modulating chronic inflammation and oxidative stress in obesity. Gal-9 is secreted out upon cell injuries, interacts with T-cell immunoglobulin-3 (Tim-3) and induces apoptosis in activated Th1 cells. Gal-9 also binds to protein disulfide isomerase (PDI), maintains PDI on surface of T cells, and increases free thiols in the disulfide/thiol cycles. To explore the molecular mechanism of obesity, we investigated Gal-9-/- and Gal-9wt/wt C57BL/6J mice fed with high fat-high sucrose (HFHS) chow. Gal-9-/- mice were resistant to diet-induced obesity associated with reduction of epididymal and mesenteric fat tissues and improved glucose tolerance compared with Gal-9wt/wt mice. However, the number of M1, M2 macrophages, and M1/M2 ratio in epididymal fat were unaltered. Under HFHS chow, Gal-9-/- mice receiving Gal-9-/- or Gal-9wt/wt bone marrow-derived cells (BMCs) demonstrated significantly lower body weight compared with Gal-9wt/wt mice receiving Gal-9-/- BMCs. We identified the binding between Gal-9 and peroxiredoxin-2 (PRDX2) in sugar chain-independent manner by nanoLC-MS/MS, immunoprecipitation, and pull-down assay. In 3T3L1 adipocytes, Gal-9 knockdown shifts PRDX2 monomer (reduced form) dominant from PRDX2 dimer (oxidized form) under oxidative stress with H2O2. The inhibition of Gal-9 in adipocytes may be a new therapeutic approach targeting the oxidative stress and subsequent glucose intolerance in obesity.


Subject(s)
Disease Susceptibility , Galectins/deficiency , Obesity/etiology , Obesity/metabolism , Oxidation-Reduction , Peroxiredoxins/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Biomarkers , Body Weight , Disease Models, Animal , Galectins/metabolism , Glucose/metabolism , Lipid Metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Protein Binding , RNA, Small Interfering/genetics
4.
J Biomed Sci ; 27(1): 24, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31937306

ABSTRACT

BACKGROUND: Galectin-9 is a ß-galactoside-binding protein with two carbohydrate recognition domains. Recent studies have revealed that galectin-9 regulates cellular biological reactions and plays a pivotal role in fibrosis. The aim of this study was to determine the role of galectin-9 in the pathogenesis of bleomycin-induced systemic sclerosis (SSc). METHODS: Human galectin-9 levels in the serum of patients with SSc and mouse sera galectin-9 levels were measured by a Bio-Plex immunoassay and enzyme-linked immunosorbent assay. Lung fibrosis was induced using bleomycin in galectin-9 wild-type and knockout mice. The effects of galectin-9 on the fibrosis markers and signaling molecules in the mouse lung tissues and primary lung fibroblast cells were assessed with western blotting and quantitative polymerase chain reaction. RESULTS: Galectin-9 levels in the serum were significantly higher (9-fold) in patients compared to those of healthy individuals. Galectin-9 deficiency in mice prominently ameliorated epithelial proliferation, collagen I accumulation, and α-smooth muscle actin expression. In addition, the galectin-9 knockout mice showed reduced protein expression levels of fibrosis markers such as Smad2/3, connective tissue growth factor, and endothelin-1. Differences between the wild-type and knockout groups were also observed in the AKT, mitogen-activated protein kinase, and c-Jun N-terminal kinase signaling pathways. Galectin-9 deficiency decreased the signal activation induced by transforming growth factor-beta in mouse primary fibroblasts, which plays a critical role in fibroblast activation and aberrant catabolism of the extracellular matrix. CONCLUSIONS: Our findings suggest that lack of galectin-9 protects against bleomycin-induced SSc. Moreover, galectin-9 might be involved in regulating the progression of fibrosis in multiple pathways.


Subject(s)
Galectins/blood , Galectins/deficiency , Pulmonary Fibrosis/drug therapy , Scleroderma, Systemic/drug therapy , Transforming Growth Factor beta/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Bleomycin/toxicity , Fibroblasts/drug effects , Lung/drug effects , Mice , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Scleroderma, Systemic/chemically induced , Signal Transduction , Smad Proteins/metabolism
5.
BMC Genomics ; 20(1): 509, 2019 Jun 18.
Article in English | MEDLINE | ID: mdl-31215398

ABSTRACT

BACKGROUND: Livestock production aims to provide meats of high and consistent eating quality. Insufficient intramuscular (IM) fat and excessive subcutaneous (SC) fat are paramount pork quality challenges. IM fat and SC fat, which are modulated by the adipogenesis of IM and SC adipocytes, play key roles in pork quality. Galectin-12 (LGALS12) was proven to be an important regulator of fat deposition in porcine. However, the current knowledge of the transcriptome-wide role of LGALS12 in adipocytes is still limited. This study was aimed to discover the different regulatory mechanisms of LGALS12 in porcine IM and SC adipocyte. RESULTS: The siRNA-mediated knockdown of the expression of LGALS12 identified 1075 and 3016 differentially expressed genes (DEGs) in IM and SC adipocytes, respectively. Among these, 585 were up- and 490 were downregulated in the IM adipocytes, while 2186 were up- and 830 were downregulated in the SC adipocytes. Moreover, 418 DGEs were observed only in the IM adipocytes, 2359 DGEs only in the SC adipocytes, and 657 DGEs in both types of adipocytes. According to Gene Ontology (GO) analysis, DEGs in both IM and SC adipocytes were mainly enriched in categories related to lipids or fat cell differentiation. Pathway analysis of the DEGs revealed 88 changed signaling pathways in the IM adipocytes and 86 in the SC adipocytes. The signaling pathways present in only one type of adipocyte were identified from among the top 50 signaling pathways in each type of adipocyte. Four signaling pathways, encompassing PI3K-AKT, cardiac muscle contraction, fatty acid metabolism and Ras, were significantly enriched in the IM adipocytes. On the other hand, four different signaling pathways, encompassing TNF, WNT, cGMP-PKG and NF-kappa B, were greatly enriched in the SC ones. The pathway changes were confirmed by chemical inhibition assays. CONCLUSIONS: Our data reveals that LGALS12 knockdown alters the expression of numerous genes involved in key biological processes in the development of adipocytes. These observations provide a global view of the role of LGALS12 in porcine IM and SC adipocytes; thus, improving our understanding of the regulatory mechanisms by which this gene acts in fat development.


Subject(s)
Adipocytes/metabolism , Galectins/genetics , Gene Expression Profiling , Muscles/cytology , Subcutaneous Fat/cytology , Swine/metabolism , Animals , Galectins/deficiency , Gene Expression Regulation/genetics , Gene Ontology , Gene Silencing , Transcription Factors/metabolism
6.
Mol Cell ; 70(1): 120-135.e8, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625033

ABSTRACT

The Ser/Thr protein kinase mTOR controls metabolic pathways, including the catabolic process of autophagy. Autophagy plays additional, catabolism-independent roles in homeostasis of cytoplasmic endomembranes and whole organelles. How signals from endomembrane damage are transmitted to mTOR to orchestrate autophagic responses is not known. Here we show that mTOR is inhibited by lysosomal damage. Lysosomal damage, recognized by galectins, leads to association of galectin-8 (Gal8) with the mTOR apparatus on the lysosome. Gal8 inhibits mTOR activity through its Ragulator-Rag signaling machinery, whereas galectin-9 activates AMPK in response to lysosomal injury. Both systems converge upon downstream effectors including autophagy and defense against Mycobacterium tuberculosis. Thus, a novel galectin-based signal-transduction system, termed here GALTOR, intersects with the known regulators of mTOR on the lysosome and controls them in response to lysosomal damage. VIDEO ABSTRACT.


Subject(s)
Autophagy , Galectins/metabolism , Lysosomes/enzymology , TOR Serine-Threonine Kinases/metabolism , Tuberculosis/enzymology , AMP-Activated Protein Kinases/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Animals , Disease Models, Animal , Female , Galectins/deficiency , Galectins/genetics , HEK293 Cells , HeLa Cells , Humans , Lysosomes/microbiology , Lysosomes/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Mycobacterium tuberculosis/pathogenicity , Signal Transduction , THP-1 Cells , TOR Serine-Threonine Kinases/genetics , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/pathology
7.
Arthritis Rheumatol ; 70(7): 1089-1101, 2018 07.
Article in English | MEDLINE | ID: mdl-29481735

ABSTRACT

OBJECTIVE: In systemic lupus erythematosus (SLE), an autoimmune disease associated with multiple organ involvement, the development of lupus nephritis determines prognosis, and arthritis impairs quality of life. Galectin 9 (Gal-9, Lgals9) is a ß-galactoside-binding lectin that has been used for clinical application in autoimmune diseases, since recombinant Gal-9, as a ligand for T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), induces apoptosis of activated CD4+TIM-3+ Th1 cells. This study was undertaken to investigate whether deficiency of Lgals9 has beneficial or deleterious effects on lupus in a murine model. METHODS: Gal-9+/+ and Gal-9-/- female BALB/c mice were injected with pristane, and the severity of arthritis, proteinuria, and levels of autoantibody production were assessed at several time points immediately following injection. At 7 months after pristane injection, renal pathologic features, the severity of joint inflammation, and formation of lipogranulomas were evaluated. Subsets of inflammatory cells in the spleen and peritoneal lavage were characterized, and expression levels of cytokines from peritoneal macrophages were analyzed. RESULTS: Lgals9 deficiency protected against the development of immune complex glomerulonephritis, arthritis, and peritoneal lipogranuloma formation in BALB/c mice in this murine model of pristane-induced lupus. The populations of T cell subsets and B cells in the spleen and peritoneum were not altered by Lgals9 deficiency in pristane-injected BALB/c mice. Furthermore, Lgals9 deficiency protected against pristane-induced lupus without altering the Toll-like receptor 7-type I interferon pathway. CONCLUSION: Gal-9 is required for the induction and development of lupus nephritis and arthritis in this murine model of SLE. The results of the current investigation provide a potential new strategy in which antagonism of Gal-9 may be beneficial for the treatment of nephritis and arthritis in patients with SLE through targeting of activated macrophages.


Subject(s)
Arthritis/genetics , Galectins/deficiency , Lupus Erythematosus, Systemic/genetics , Lupus Nephritis/genetics , Animals , B-Lymphocytes/physiology , Disease Models, Animal , Female , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Terpenes , Th1 Cells/physiology
8.
mBio ; 8(4)2017 07 25.
Article in English | MEDLINE | ID: mdl-28743815

ABSTRACT

Group A streptococcus (GAS) is an important human pathogen that causes a wide variety of cutaneous and systemic infections. Although originally thought to be an extracellular bacterium, numerous studies have demonstrated that GAS can trigger internalization into nonimmune cells to escape from immune surveillance or antibiotic-mediated killing. Epithelial cells possess a defense mechanism involving autophagy-mediated targeting and killing of GAS within lysosome-fused autophagosomes. In endothelial cells, in contrast, we previously showed that autophagy is not sufficient for GAS killing. In the present study, we showed higher galectin-3 (Gal-3) expression and lower Gal-8 expression in endothelial cells than in epithelial cells. The recruitment of Gal-3 to GAS is higher and the recruitment of Gal-8 to GAS is lower in endothelial cells than in epithelial cells. We further showed that Gal-3 promotes GAS replication and diminishes the recruitment of Gal-8 and ubiquitin, the latter of which is a critical protein for autophagy sequestration. After knockdown of Gal-3 in endothelial cells, the colocalization of Gal-8, parkin, and ubiquitin-decorated GAS is significantly increased, as is the interaction of Gal-8 and parkin, an E3 ligase. Furthermore, inhibition of Gal-8 in epithelial cells attenuates recruitment of parkin; both Gal-8 and parkin contribute to ubiquitin recruitment and GAS elimination. Animal studies confirmed that Gal-3-knockout mice develop less-severe skin damage and that GAS replication can be detected only in the air pouch and not in organs and endothelial cells. These results demonstrate that Gal-3 inhibits ubiquitin recruitment by blocking Gal-8 and parkin recruitment, resulting in GAS replication in endothelial cells.IMPORTANCE In epithelial cells, GAS can be efficiently killed within the lysosome-fused autophaosome compartment. However, we previously showed that, in spite of LC-3 recruitment, the autophagic machinery is not sufficient for GAS killing in endothelial cells. In this report, we provide the first evidence that Gal-3, highly expressed in endothelial cells, blocks the tagging of ubiquitin to GAS by inhibiting recruitment of Gal-8 and parkin, leading to an enhancement of GAS replication. We also provide the first demonstration that Gal-8 can interact with parkin, the critical E3 ligase, for resistance to intracellular bacteria by facilitating the decoration of bacteria with ubiquitin chains. Our findings reveal that differential levels of Gal-3 and Gal-8 expression and recruitment to GAS between epithelial cells and endothelial cells may contribute to the different outcomes of GAS elimination or survival and growth of GAS in these two types of cells.


Subject(s)
Galectin 3/metabolism , Galectins/metabolism , Streptococcus pyogenes/metabolism , Ubiquitin-Protein Ligases/metabolism , A549 Cells , Animals , Autophagy , Blood Proteins , Endothelial Cells/microbiology , Epithelial Cells/microbiology , Galectin 3/deficiency , Galectin 3/genetics , Galectins/antagonists & inhibitors , Galectins/deficiency , Galectins/genetics , Gene Silencing , Humans , Mice , Mice, Knockout , RNA Interference , Skin/microbiology , Skin/pathology , Streptococcus pyogenes/growth & development , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Genetics ; 199(1): 247-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398792

ABSTRACT

Clinically relevant features of monogenic diseases, including severity of symptoms and age of onset, can vary widely in response to environmental differences as well as to the presence of genetic modifiers affecting the trait's penetrance and expressivity. While a better understanding of modifier loci could lead to treatments for Mendelian diseases, the rarity of individuals harboring both a disease-causing allele and a modifying genotype hinders their study in human populations. We examined the genetic architecture of monogenic trait modifiers using a well-characterized yeast model of the human Mendelian disease classic galactosemia. Yeast strains with loss-of-function mutations in the yeast ortholog (GAL7) of the human disease gene (GALT) fail to grow in the presence of even small amounts of galactose due to accumulation of the same toxic intermediates that poison human cells. To isolate and individually genotype large numbers of the very rare (∼0.1%) galactose-tolerant recombinant progeny from a cross between two gal7Δ parents, we developed a new method, called "FACS-QTL." FACS-QTL improves upon the currently used approaches of bulk segregant analysis and extreme QTL mapping by requiring less genome engineering and strain manipulation as well as maintaining individual genotype information. Our results identified multiple distinct solutions by which the monogenic trait could be suppressed, including genetic and nongenetic mechanisms as well as frequent aneuploidy. Taken together, our results imply that the modifiers of monogenic traits are likely to be genetically complex and heterogeneous.


Subject(s)
Aneuploidy , Genes, Modifier , Genetic Variation , Quantitative Trait Loci , Saccharomyces cerevisiae/genetics , Alleles , Chromosome Mapping/methods , Galactose/metabolism , Galectins/deficiency , Galectins/genetics
10.
Methods Mol Biol ; 1207: 133-51, 2015.
Article in English | MEDLINE | ID: mdl-25253138

ABSTRACT

Numerous protocols exist for investigating leukocyte recruitment both in vitro and in vivo. Here we describe three of these methods; an in vitro flow chamber assay, intravital microscopy, and zymosan-induced peritonitis, and give details as to how they can be used to study the actions of galectins on this crucial process.


Subject(s)
Cell Movement , Galectins/metabolism , Leukocytes/cytology , Animals , Cell Culture Techniques , Cell Movement/drug effects , Cell Separation , Galectins/deficiency , Galectins/genetics , Gene Knockout Techniques , Human Umbilical Vein Endothelial Cells/cytology , Humans , Image Processing, Computer-Assisted , Leukocytes/immunology , Male , Mice , Microscopy , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/metabolism , Zymosan/pharmacology
11.
Methods Mol Biol ; 1207: 327-41, 2015.
Article in English | MEDLINE | ID: mdl-25253151

ABSTRACT

Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role(s) of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are ß-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), which comprise an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies.


Subject(s)
Galectins/genetics , Gene Expression Regulation/genetics , Gene Knockdown Techniques/methods , Gene Knockout Techniques/methods , Zebrafish/genetics , Animals , Base Sequence , Embryo, Nonmammalian , Female , Galectins/deficiency , Injections , Male , Morpholinos/genetics , Phenotype , RNA/genetics , Zebrafish/embryology
12.
PLoS One ; 8(8): e72488, 2013.
Article in English | MEDLINE | ID: mdl-23967307

ABSTRACT

Human monocytes/macrophages (M/M(Ф)) of the innate immunity sense and respond to microbial products via specific receptor coupling with stimulatory (such as TLR) and inhibitory (such as Tim-3) receptors. Current models imply that Tim-3 expression on M/M(Ø) can deliver negative signaling to TLR-mediated IL-12 expression through trans association with its ligand Galectin-9 (Gal-9) presented by other cells. However, Gal-9 is also expressed within M/M(Ø), and the effect of intracellular Gal-9 on Tim-3 activities and inflammatory responses in the same M/M(Ø) remains unknown. In this study, our data suggest that Tim-3 and IL-12/IL-23 gene transcriptions are regulated by enhanced or silenced Gal-9 expression within monocytes through synergizing with TLR signaling. Additionally, TLR activation facilitates Gal-9/Tim-3 cis association within the same M/M(Ø) to differentially regulate IL-12/IL-23 expressions through STAT-3 phosphorylation. These results reveal a ligand (Gal-9) compartment-dependent regulatory effect on receptor (Tim-3) activities and inflammatory responses via TLR pathways--a novel mechanism underlying cellular responses to external or internal cues.


Subject(s)
Galectins/metabolism , Gene Expression Regulation , Interleukin-12/genetics , Interleukin-23/genetics , Membrane Proteins/metabolism , Monocytes/cytology , Toll-Like Receptors/metabolism , Cell Line , Galectins/deficiency , Galectins/genetics , Gene Silencing , Hepatitis A Virus Cellular Receptor 2 , Humans , Intracellular Space/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic
13.
J Biol Chem ; 288(33): 23776-87, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23836896

ABSTRACT

Demyelination and axonal damage in multiple sclerosis (MS) are thought to be a consequence of inflammatory processes that are perpetuated by activated glia and infiltrating leukocytes. Galectin-9 is a ß-galactoside binding lectin capable of modulating immune responses and appears to be up-regulated in MS. However, its role in the pathogenesis of MS has yet to be determined. Here, we report that proinflammatory cytokines induce galectin-9 (Gal-9) expression in primary astrocytes and the mechanism by which TNF up-regulates Gal-9. Astrocytes did not express Gal-9 under basal conditions nor did IL-6, IL-10, or IL-13 trigger Gal-9 expression. In contrast, IL-1ß, IFN-γ, and particularly TNF up-regulated Gal-9 in astrocytes. TNF-induced Gal-9 expression was dependent on TNF receptor 1 (TNFR1) as TNF failed to induce Gal-9 in TNFR1(-/-) astrocytes. Blockade of the JNK MAP kinase pathway with the JNK inhibitor SP600125 abrogated TNF-induced Gal-9, whereas p38 and MEK inhibitors had minimal effects. Furthermore, specific knockdown of c-Jun via siRNA in astrocytes before TNF treatment greatly suppressed Gal-9 transcription, suggesting that TNF induces astroglial Gal-9 through the TNF/TNFR1/JNK/cJun signaling pathway. Finally, utilizing astrocytes from Lgals9 mutant (Gal-9(-/-)) mice as well as a myelin basic protein-specific Tim-3(+) encephalitogenic T-cell clone (LCN-8), we found that conditioned medium from TNF-stimulated Gal-9(+/+) but not Gal-9(-/-) astrocytes increased the percentage of apoptotic encephalitogenic T-cells. Together, our results suggest that Gal-9 is induced in astrocytes by TNF via the JNK/c-Jun pathway and that astrocyte-derived Gal-9 may function as an immunoregulatory protein in response to ongoing neuroinflammation.


Subject(s)
Apoptosis/drug effects , Astrocytes/metabolism , Encephalitis/pathology , Galectins/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Animals , Apoptosis/genetics , Astrocytes/drug effects , Astrocytes/pathology , Cells, Cultured , Enzyme Activation/drug effects , Galectins/deficiency , Galectins/genetics , Inflammation Mediators/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/metabolism , Sus scrofa , T-Lymphocytes/drug effects , Transcription, Genetic/drug effects , Up-Regulation/genetics
14.
Cell Death Dis ; 4: e496, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23412389

ABSTRACT

Ras proteins undergo an incompletely understood trafficking process in the cell. Rasosomes are protein nanoparticles of 80-100 nm diameter that carry lipidated Ras isoforms (H-Ras and N-Ras) as well as their effectors through the cytoplasm and near the plasma membrane (PM). In this study, we identified the subcellular origin of rasosomes and how they spread Ras proteins through the cell. We found no dependency of rasosome formation on galectins, or on the GDP-/GTP-bound state of Ras. We found that significantly more rasosomes are associated with forms of Ras that are localized to the Golgi, namely N-Ras or the singly palmitoylated H-Ras mutant (C181S). To explore the possibility that rasosome originate from the Golgi, we used photoactivatable (PA)-GFP-H-Ras mutants and showed that rasosomes bud from the Golgi in a two-step mechanism. Newly released rasosomes first move in an energy-dependent directed fashion and then convert to randomly diffusing rasosomes. Dual fluorescence time-lapse imaging revealed the appearance of dually labeled rasosomes, indicating a dynamic exchange of cytoplasmic and PM-associated Ras with rasosome-associated Ras. Finally, higher levels of rasosomes correlate with higher levels of ERK phosphorylation, a key marker of Ras downstream signaling. We suggest that H-Ras and N-Ras proteins exchange with rasosomes that can function as carriers of palmitoylated Ras and its signals.


Subject(s)
Golgi Apparatus/metabolism , ras Proteins/metabolism , Animals , COS Cells , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/metabolism , Galectins/deficiency , Galectins/genetics , Galectins/metabolism , Golgi Apparatus/genetics , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Lipoylation , Mice , Mutation , Nanoparticles/chemistry , Phosphorylation , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Time-Lapse Imaging , Transfection
15.
Expert Rev Mol Med ; 10: e17, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18549522

ABSTRACT

Galectins are a family of animal lectins that bind beta-galactosides. Outside the cell, galectins bind to cell-surface and extracellular matrix glycans and thereby affect a variety of cellular processes. However, galectins are also detectable in the cytosol and nucleus, and may influence cellular functions such as intracellular signalling pathways through protein-protein interactions with other cytoplasmic and nuclear proteins. Current research indicates that galectins play important roles in diverse physiological and pathological processes, including immune and inflammatory responses, tumour development and progression, neural degeneration, atherosclerosis, diabetes, and wound repair. Some of these have been discovered or confirmed by using genetically engineered mice deficient in a particular galectin. Thus, galectins may be a therapeutic target or employed as therapeutic agents for inflammatory diseases, cancers and several other diseases.


Subject(s)
Galectins , Animals , Atherosclerosis/physiopathology , Drug Screening Assays, Antitumor , Galactosides/metabolism , Galectins/chemistry , Galectins/deficiency , Galectins/physiology , Galectins/therapeutic use , Gene Expression , Humans , Immune System/physiology , Inflammation/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Neoplasm Proteins/physiology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Polysaccharides/metabolism , Signal Transduction/physiology , Structure-Activity Relationship , Subcellular Fractions/metabolism , Substrate Specificity
16.
Glycobiology ; 16(1): 36-45, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16166603

ABSTRACT

Galectin 3 belongs to a family of glycoconjugate-binding proteins that participate in cellular homeostasis by modulating cell growth, adhesion, and signaling. We studied adult galectin 3 null mutant (Gal 3-/-) and wild-type (WT) mice to gain insights into the role of galectin 3 in the kidney. By immunofluorescence, galectin 3 was found in collecting duct (CD) principal and intercalated cells in some regions of the kidney, as well as in the thick ascending limbs at lower levels. Compared to WT mice, Gal 3-/- mice had approximately 11% fewer glomeruli (p < 0.04), associated with kidney hypertrophy (p < 0.006). In clearance experiments, urinary chloride excretion was found to be higher in Gal 3-/- than in WT mice (p < 0.04), but there was no difference in urinary bicarbonate excretion, in glomerular filtration, or urinary flow rates. Under chronic low sodium diet, Gal 3-/- mice had lower extracellular fluid (ECF) volume than WT mice (p < 0.05). Plasma aldosterone concentration was higher in Gal 3-/- than in WT mice (p < 0.04), which probably caused the observed increase in alpha-epithelial sodium channel (alpha-ENaC) protein abundance in the mutant mice (p < 0.001). Chronic high sodium diet resulted paradoxically in lower blood pressure (p < 0.01) in Gal 3-/- than in WT. We conclude that Gal 3-/- mice have mild renal chloride loss, which causes chronic ECF volume contraction and reduced blood pressure levels.


Subject(s)
Galectins/metabolism , Glomerular Filtration Rate/physiology , Homeostasis/physiology , Kidney Tubules, Collecting/metabolism , Nuclear Proteins/metabolism , Animals , Biological Transport, Active/physiology , Galectin 3 , Galectins/deficiency , Kidney Tubules, Collecting/ultrastructure , Mice , Mice, Mutant Strains , Nuclear Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...