Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
J Gastroenterol Hepatol ; 35(12): 2158-2169, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32424849

ABSTRACT

BACKGROUND AND AIM: Galectin-1 plays a protective role against colitis by binding with polylactosamine structures on macrophages in ß-1,4-galactosyltransferase I-deficient mice, but the precise function of galectin-1 remains unknown. In the present study, we investigated the anti-inflammatory role of galectin-1 on macrophages to ameliorate ulcerative colitis in both animal model and human tissue samples. METHODS: The expression of galectin-1 in colonic tissues of ulcerative colitis patients was evaluated by immunohistochemistry. Cytokine production of mouse bone marrow-derived macrophages (BMDMs) cultured with galectin-1 was investigated. Galectin-1 binding capacity and polylactosamine expression in macrophages stimulated with lipopolysaccharides were evaluated by flow cytometry. BMDMs cultured with galectin-1 were transferred into Recombination activating gene (Rag) 2-/- mice, and the severity of the dextran sodium sulfate-induced colitis model was investigated. Furthermore, RNA sequencing was performed to characterize macrophages treated with galectin-1. RESULTS: In ulcerative colitis patients, tissue expression of galectin-1was decreased in inflamed mucosa compared with non-inflamed mucosa. Galectin-1 induced interleukin-10 production in BMDMs, and the interleukin-10 production was abrogated by lactose, which inhibits the interaction of oligosaccharide-galectin binding. Dextran sodium sulfate colitis was significantly ameliorated in Rag2-/- mice undergoing galectin-1-treated BMDM transfer compared with those undergoing vehicle-treated BMDM transfer. RNA sequencing revealed that treatment with galectin-1 increased the expression of CCAAT/enhancer binding protein ß and CD163, but decreased the expression of CD80 on BMDMs. CONCLUSION: Galectin-1, whose expression is decreased in the inflamed mucosa of ulcerative colitis patients, can ameliorate murine colitis by conferring oligosaccharide-dependent anti-inflammatory properties to macrophages.


Subject(s)
Colitis, Ulcerative/genetics , Galectin 1/physiology , Gene Expression , Macrophages/metabolism , Macrophages/physiology , Oligosaccharides , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Colitis, Ulcerative/drug therapy , Disease Models, Animal , Galectin 1/genetics , Galectin 1/metabolism , Galectin 1/therapeutic use , Humans , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Lactose/pharmacology , Mice, Inbred C57BL , Oligosaccharides/metabolism , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
2.
Proc Natl Acad Sci U S A ; 117(12): 6630-6639, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32161138

ABSTRACT

Aging elicits quantitative and qualitative changes in different immune components, leading to disruption of tolerogenic circuits and development of autoimmune disorders. Galectin-1 (Gal1), an endogenous glycan-binding protein, has emerged as a regulator of immune cell homeostasis by shaping the fate of myeloid and lymphoid cells. Here, we demonstrate that aged Gal1-null mutant (Lgals1-/- ) mice develop a spontaneous inflammatory process in salivary glands that resembles Sjögren's syndrome. This spontaneous autoimmune phenotype was recapitulated in mice lacking ß1,6N-acetylglucosaminyltransferase V (Mgat5), an enzyme responsible for generating ß1,6-branched complex N-glycans, which serve as a major ligand for this lectin. Lack of Gal1 resulted in CD11c+ dendritic cells (DCs) with higher immunogenic potential, lower frequency of Foxp3+ regulatory T cells (Tregs), and increased number of CD8+ T cells with greater effector capacity. Supporting its tolerogenic activity, Gal1 expression decreased with age in autoimmunity-prone nonobese diabetic (NOD) mice. Treatment with recombinant Gal1 restored tolerogenic mechanisms and reduced salivary gland inflammation. Accordingly, labial biopsies from primary Sjögren's syndrome patients showed reduced Gal1 expression concomitant with higher number of infiltrating CD8+ T cells. Thus, endogenous Gal1 serves as a homeostatic rheostat that safeguards immune tolerance and prevents age-dependent development of spontaneous autoimmunity.


Subject(s)
Autoimmune Diseases/pathology , Galectin 1/physiology , Immune Tolerance/immunology , Salivary Glands/pathology , Sialadenitis/pathology , Sjogren's Syndrome/pathology , T-Lymphocytes, Regulatory/immunology , Adult , Age Factors , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Case-Control Studies , Dendritic Cells/immunology , Female , Glycosylation , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Middle Aged , N-Acetylglucosaminyltransferases/physiology , Polysaccharides/metabolism , Salivary Glands/immunology , Salivary Glands/metabolism , Sialadenitis/immunology , Sialadenitis/metabolism , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism
3.
J Clin Invest ; 129(12): 5553-5567, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31710313

ABSTRACT

Immune checkpoint inhibitors (ICIs), although promising, have variable benefit in head and neck cancer (HNC). We noted that tumor galectin-1 (Gal1) levels were inversely correlated with treatment response and survival in patients with HNC who were treated with ICIs. Using multiple HNC mouse models, we show that tumor-secreted Gal1 mediates immune evasion by preventing T cell migration into the tumor. Mechanistically, Gal1 reprograms the tumor endothelium to upregulate cell-surface programmed death ligand 1 (PD-L1) and galectin-9. Using genetic and pharmacological approaches, we show that Gal1 blockade increases intratumoral T cell infiltration, leading to a better response to anti-PD1 therapy with or without radiotherapy. Our study reveals the function of Gal1 in transforming the tumor endothelium into an immune-suppressive barrier and that its inhibition synergizes with ICIs.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Endothelium/physiology , Galectin 1/physiology , Head and Neck Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , B7-H1 Antigen/physiology , Female , Galectin 1/antagonists & inhibitors , Galectins/physiology , Head and Neck Neoplasms/immunology , Humans , Immune Tolerance , Immunotherapy , Male , Mice , Mice, Inbred C57BL , Middle Aged , STAT1 Transcription Factor/physiology
4.
FASEB J ; 33(7): 7995-8007, 2019 07.
Article in English | MEDLINE | ID: mdl-30897344

ABSTRACT

Chronic liver inflammation (CLI) is a risk factor for development of hepatocellular carcinoma (HCC). Galectin-1 (Gal1) is involved in the regulation of inflammation, angiogenesis, and tumorigenesis, exhibiting multiple anti-inflammatory and protumorigenic activities. We aimed to explore its regulatory role in CLI and HCC progression using an established model of CLI-mediated HCC development, Abcb4 [multidrug-resistance 2 (Mdr2)]-knockout (KO) mice, which express high levels of Gal1 in the liver. We generated double-KO (dKO) Gal1-KO/Mdr2-KO mice on C57BL/6 and FVB/N genetic backgrounds and compared HCC development in the generated strains with their parental Mdr2-KO strains. Loss of Gal1 increased liver injury, inflammation, fibrosis, and ductular reaction in dKO mice of both strains starting from an early age. Aged dKO mutants displayed earlier hepatocarcinogenesis and increased tumor size compared with control Mdr2-KO mice. We found that osteopontin, a well-known modulator of HCC development, and oncogenic proteins Ntrk2 (TrkB) and S100A4 were overexpressed in dKO compared with Mdr2-KO livers. Our results demonstrate that in Mdr2-KO mice, a model of CLI-mediated HCC, Gal1-mediated protection from hepatitis, liver fibrosis, and HCC initiation dominates over its known procarcinogenic activities at later stages of HCC development. These findings suggest that anti-Gal1 treatments may not be applicable at all stages of CLI-mediated HCC.-Potikha, T., Pappo, O., Mizrahi, L., Olam, D., Maller, S. M., Rabinovich, G. A., Galun, E., Goldenberg, D. S. Lack of galectin-1 exacerbates chronic hepatitis, liver fibrosis, and carcinogenesis in murine hepatocellular carcinoma model.


Subject(s)
Galectin 1/physiology , Hepatitis/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Precancerous Conditions/metabolism , Alternative Splicing , Animals , Cell Division , Chronic Disease , Cocarcinogenesis , Female , Galectin 1/deficiency , Galectin 1/genetics , Hep G2 Cells , Hepatitis/genetics , Hepatitis/pathology , Hepatocytes/pathology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Neoplasm Proteins/genetics , Osteopontin/biosynthesis , Osteopontin/deficiency , Osteopontin/genetics , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Specific Pathogen-Free Organisms
5.
FASEB J ; 33(5): 5836-5849, 2019 05.
Article in English | MEDLINE | ID: mdl-30753093

ABSTRACT

We have established early-gestation chorionic villus-derived placenta mesenchymal stromal cells (PMSCs) as a potential treatment for spina bifida (SB), a neural tube defect. Our preclinical studies demonstrated that PMSCs have the potential to cure hind limb paralysis in the fetal lamb model of SB via a paracrine mechanism. PMSCs exhibit neuroprotective function by increasing cell number and neurites, as shown by indirect coculture and direct addition of PMSC-conditioned medium to the staurosporine-induced apoptotic human neuroblastoma cell line, SH-SY5Y. PMSC-conditioned medium suppressed caspase activity in apoptotic SH-SY5Y cells, suggesting that PMSC secretome contributes to neuronal survival after injury. As a part of PMSC secretome, PMSC exosomes were isolated and extensively characterized; their addition to apoptotic SH-SY5Y cells mediated an increase in neurites, suggesting that they exhibit neuroprotective function. Proteomic and RNA sequencing analysis revealed that PMSC exosomes contain several proteins and RNAs involved in neuronal survival and development. Galectin 1 was highly expressed on the surface of PMSCs and PMSC exosomes. Preincubation of exosomes with anti-galectin 1 antibody decreased their neuroprotective effect, suggesting that PMSC exosomes likely impart their effect via binding of galectin 1 to cells. Future studies will include in-depth analyses of the role of PMSC exosomes on neuroprotection and their clinical applications.-Kumar, P., Becker, J. C., Gao, K., Carney, R. P., Lankford, L., Keller, B. A., Herout, K., Lam, K. S., Farmer, D. L., Wang, A. Neuroprotective effect of placenta-derived mesenchymal stromal cells: role of exosomes.


Subject(s)
Mesenchymal Stem Cells/cytology , Placenta/cytology , Spinal Dysraphism/therapy , Stromal Cells/cytology , Animals , Apoptosis , Cattle , Cell Line, Tumor , Coculture Techniques , Culture Media, Conditioned/chemistry , Exosomes/metabolism , Female , Galectin 1/physiology , Humans , Mesenchymal Stem Cell Transplantation , Mesoderm/cytology , Neural Tube Defects/therapy , Neurites/metabolism , Oxidative Stress , Pregnancy , Sheep , Signal Transduction , Staurosporine
6.
FASEB J ; 33(2): 2498-2513, 2019 02.
Article in English | MEDLINE | ID: mdl-30277820

ABSTRACT

VEGFA and TGF-ß are known major angiogenic and fibrogenic factors. Galectin-1, encoded by lectin, galactoside-binding, soluble ( LGALS) 1, has attracted growing attention for its facilitatory role in angiogenesis and fibrosis through its modification of VEGFA and TGF-ß receptor signaling pathways. We reveal galectin-1 involvement in the mouse model of laser-induced choroidal neovascularization (CNV) and subretinal fibrosis, both of which represent the pathogenesis of age-related macular degeneration (AMD). Neither deletion nor overexpression of Lgals1 affected physiologic retinal development or visual function. Galectin-1/ Lgals1 was upregulated by CNV induction, whereas deletion of Lgals1 suppressed CNV together with downstream molecules of VEGF receptor (VEGFR)2. Loss of Lgals1 also attenuated subretinal fibrosis, expression of epithelial-mesenchymal transition (EMT) markers including Snai1, and phosphorylation of SMAD family member 2. Supporting these in vivo findings, silencing of LGALS1 in human retinal pigment epithelial (RPE) cells inhibited TGF-ß1-induced EMT-related molecules and cell motilities. Conversely, overexpression of Lgals1 enhanced CNV and subretinal fibrosis. Specimens from patients with AMD demonstrated colocalization of galectin-1 with VEGFR2 in neovascular endothelial cells and with phosphorylated SMAD2 in RPE cells. These results suggested a biologic significance of galectin-1 as a key promotor for both angiogenesis and fibrosis in eyes with AMD.-Wu, D., Kanda, A., Liu, Y., Kase, S., Noda, K., Ishida, S. Galectin-1 promotes choroidal neovascularization and subretinal fibrosis mediated via epithelial-mesenchymal transition.


Subject(s)
Choroidal Neovascularization/pathology , Epithelial-Mesenchymal Transition , Fibrosis/pathology , Galectin 1/physiology , Macular Degeneration/pathology , Retina/pathology , Retinal Pigment Epithelium/pathology , Aged, 80 and over , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Disease Models, Animal , Female , Fibrosis/genetics , Fibrosis/metabolism , Humans , Laser Coagulation/adverse effects , Macular Degeneration/genetics , Macular Degeneration/metabolism , Male , Mice , Mice, Inbred C57BL , Retina/metabolism , Retinal Pigment Epithelium/metabolism
7.
FASEB J ; 33(1): 373-387, 2019 01.
Article in English | MEDLINE | ID: mdl-29975570

ABSTRACT

Chronic exposure of tubular renal cells to high glucose contributes to tubulointerstitial changes in diabetic nephropathy. In the present study, we identified a new fibrosis gene called galectin-1 (Gal-1), which is highly expressed in tubular cells of kidneys of type 1 and type 2 diabetic mouse models. Gal-1 protein and mRNA expression showed significant increase in kidney cortex of heterozygous Akita+/- and db/db mice compared with wild-type mice. Mouse proximal tubular cells exposed to high glucose showed significant increase in phosphorylation of Akt and Gal-1. We cloned Gal-1 promoter and identified the transcription factor AP4 as binding to the Gal-1 promoter to up-regulate its function. Transfection of cells with plasmid carrying mutations in the binding sites of AP4 to Gal-1 promoter resulted in decreased protein function of Gal-1. In addition, inhibition of Gal-1 by OTX-008 showed significant decrease in p-Akt/AP4 and protein-promoter activity of Gal-1 and fibronectin. Moreover, down-regulation of AP4 by small interfering RNA resulted in a significant decrease in protein expression and promoter activity of Gal-1. We found that kidney of Gal-1-/- mice express very low levels of fibronectin protein. In summary, Gal-1 is highly expressed in kidneys of type 1 and 2 diabetic mice, and AP4 is a major transcription factor that activates Gal-1 under hyperglycemia. Inhibition of Gal-1 by OTX-008 blocks activation of Akt and prevents accumulation of Gal-1, suggesting a novel role of Gal-1 inhibitor as a possible therapeutic target to treat renal fibrosis in diabetes.-Al-Obaidi, N., Mohan, S., Liang, S., Zhao, Z., Nayak, B. K., Li, B., Sriramarao, P., Habib, S. L. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Fibrosis/metabolism , Galectin 1/physiology , Gene Expression Regulation/drug effects , Kidney Tubules, Proximal/metabolism , Animals , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Fibronectins/metabolism , Fibrosis/etiology , Fibrosis/pathology , Glucose/administration & dosage , HEK293 Cells , Humans , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Promoter Regions, Genetic
8.
Proc Natl Acad Sci U S A ; 115(16): E3769-E3778, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29615514

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) remains one of the most lethal tumor types, with extremely low survival rates due to late diagnosis and resistance to standard therapies. A more comprehensive understanding of the complexity of PDA pathobiology, and especially of the role of the tumor microenvironment in disease progression, should pave the way for therapies to improve patient response rates. In this study, we identify galectin-1 (Gal1), a glycan-binding protein that is highly overexpressed in PDA stroma, as a major driver of pancreatic cancer progression. Genetic deletion of Gal1 in a Kras-driven mouse model of PDA (Ela-KrasG12Vp53-/- ) results in a significant increase in survival through mechanisms involving decreased stroma activation, attenuated vascularization, and enhanced T cell infiltration leading to diminished metastasis rates. In a human setting, human pancreatic stellate cells (HPSCs) promote cancer proliferation, migration, and invasion via Gal1-driven pathways. Moreover, in vivo orthotopic coinjection of pancreatic tumor cells with Gal1-depleted HPSCs leads to impaired tumor formation and metastasis in mice. Gene-expression analyses of pancreatic tumor cells exposed to Gal1 reveal modulation of multiple regulatory pathways involved in tumor progression. Thus, Gal1 hierarchically regulates different events implicated in PDA biology including tumor cell proliferation, invasion, angiogenesis, inflammation, and metastasis, highlighting the broad therapeutic potential of Gal1-specific inhibitors, either alone or in combination with other therapeutic modalities.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Galectin 1/physiology , Galectins/physiology , Molecular Targeted Therapy , Pancreatic Neoplasms/therapy , Animals , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Cell Division/genetics , Cell Movement/genetics , Culture Media, Conditioned , Galectins/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Ontology , Heterografts , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Metastasis , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/transplantation , Paracrine Communication , RNA, Small Interfering/genetics , Stromal Cells/metabolism , Tumor Microenvironment
9.
Sci Rep ; 8(1): 2725, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426942

ABSTRACT

Galectin-1 (Gal-1) is required for the development of B cells in the bone marrow (BM), however very little is known about the contribution of Gal-1 to the development of B cell regulatory function. Here, we report an important role for Gal-1 in the induction of B cells regulatory function. Mice deficient of Gal-1 (Gal-1-/-) showed significant loss of Transitional-2 (T2) B cells, previously reported to include IL-10+ regulatory B cells. Gal-1-/- B cells stimulated in vitro via CD40 molecules have impaired IL-10 and Tim-1 expression, the latter reported to be required for IL-10 production in regulatory B cells, and increased TNF-α expression compared to wild type (WT) B cells. Unlike their WT counterparts, T2 and T1 Gal-1-/- B cells did not suppress TNF-α expression by CD4+ T cells activated in vitro with allogenic DCs (allo-DCs), nor were they suppressive in vivo, being unable to delay MHC-class I mismatched skin allograft rejection following adoptive transfer. Moreover, T cells stimulated with allo-DCs show an increase in their survival when co-cultured with Gal-1-/- T2 and MZ B cells compared to WT T2 and MZ B cells. Collectively, these data suggest that Gal-1 contributes to the induction of B cells regulatory function.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Dendritic Cells/immunology , Galectin 1/physiology , Graft Survival , Lymphocyte Activation , Allografts , Animals , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/pathology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Skin Transplantation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
11.
Proc Natl Acad Sci U S A ; 113(33): E4820-7, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27496330

ABSTRACT

Branching morphogenesis in the mammary gland is achieved by the migration of epithelial cells through a microenvironment consisting of stromal cells and extracellular matrix (ECM). Here we show that galectin-1 (Gal-1), an endogenous lectin that recognizes glycans bearing N-acetyllactosamine (LacNAc) epitopes, induces branching migration of mammary epithelia in vivo, ex vivo, and in 3D organotypic cultures. Surprisingly, Gal-1's effects on mammary patterning were independent of its glycan-binding ability and instead required localization within the nuclei of mammary epithelia. Nuclear translocation of Gal-1, in turn, was regulated by discrete cell-surface glycans restricted to the front of the mammary end buds. Specifically, α2,6-sialylation of terminal LacNAc residues in the end buds masked Gal-1 ligands, thereby liberating the protein for nuclear translocation. Within mammary epithelia, Gal-1 localized within nuclear Gemini bodies and drove epithelial invasiveness. Conversely, unsialylated LacNAc glycans, enriched in the epithelial ducts, sequestered Gal-1 in the extracellular environment, ultimately attenuating invasive potential. We also found that malignant breast cells possess higher levels of nuclear Gal-1 and α2,6-SA and lower levels of LacNAc than nonmalignant cells in culture and in vivo and that nuclear localization of Gal-1 promotes a transformed phenotype. Our findings suggest that differential glycosylation at the level of tissue microanatomy regulates the nuclear function of Gal-1 in the context of mammary gland morphogenesis and in cancer progression.


Subject(s)
Cell Nucleus/metabolism , Galectin 1/physiology , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Animal/etiology , Morphogenesis , Polysaccharides/physiology , Animals , Female , Glycosylation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
Proc Natl Acad Sci U S A ; 113(33): E4837-46, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27457925

ABSTRACT

Galectin-1 (Gal-1), a glycan-binding protein with broad antiinflammatory activities, functions as a proresolving mediator in autoimmune and chronic inflammatory disorders. However, its role in allergic airway inflammation has not yet been elucidated. We evaluated the effects of Gal-1 on eosinophil function and its role in a mouse model of allergic asthma. Allergen exposure resulted in airway recruitment of Gal-1-expressing inflammatory cells, including eosinophils, as well as increased Gal-1 in extracellular spaces in the lungs. In vitro, extracellular Gal-1 exerted divergent effects on eosinophils that were N-glycan- and dose-dependent. At concentrations ≤0.25 µM, Gal-1 increased eosinophil adhesion to vascular cell adhesion molecule-1, caused redistribution of integrin CD49d to the periphery and cell clustering, but inhibited ERK(1/2) activation and eotaxin-1-induced migration. Exposure to concentrations ≥1 µM resulted in ERK(1/2)-dependent apoptosis and disruption of the F-actin cytoskeleton. At lower concentrations, Gal-1 did not alter expression of adhesion molecules (CD49d, CD18, CD11a, CD11b, L-selectin) or of the chemokine receptor CCR3, but decreased CD49d and CCR3 was observed in eosinophils treated with higher concentrations of this lectin. In vivo, allergen-challenged Gal-1-deficient mice exhibited increased recruitment of eosinophils and CD3(+) T lymphocytes in the airways as well as elevated peripheral blood and bone marrow eosinophils relative to corresponding WT mice. Further, these mice had an increased propensity to develop airway hyperresponsiveness and displayed significantly elevated levels of TNF-α in lung tissue. This study suggests that Gal-1 can limit eosinophil recruitment to allergic airways and suppresses airway inflammation by inhibiting cell migration and promoting eosinophil apoptosis.


Subject(s)
Asthma/etiology , Eosinophilia/etiology , Galectin 1/physiology , Animals , Apoptosis , Cell Adhesion , Chemokines/analysis , Cytokines/analysis , Eosinophils/physiology , Galectin 1/analysis , Lung/chemistry , Mice , Mice, Inbred C57BL
13.
Biol Res ; 49(1): 33, 2016 Jul 27.
Article in English | MEDLINE | ID: mdl-27459991

ABSTRACT

BACKGROUND: Glioblastoma is one of the most aggressive cancers of the brain. Malignant traits of glioblastoma cells include elevated migration, proliferation and survival capabilities. Galectins are unconventionally secreted glycan-binding proteins that modulate processes of cell adhesion, migration, proliferation and apoptosis by interacting with beta-galactosides of cell surface glycoproteins and the extracellular matrix. Galectin-8 is one of the galectins highly expressed in glioblastoma cells. It has a unique selectivity for terminally sialylated glycans recently found enhanced in these highly malignant cells. A previous study in glioblastoma cell lines reported that Gal-8 coating a plastic surface stimulates two-dimensional motility. Because in other cells Gal-8 arrests proliferation and induces apoptosis, here we extend its study by analyzing all of these processes in a U87 glioblastoma cell model. METHODS: We used immunoblot and RT-PCR for Gal-8 expression analysis, recombinant Gal-8 produced in a bacteria system for Gal-8 treatment of the cells, and shRNA in lentivirus transduction for Gal-8 silencing. Cell migration as assessed in transwell filters. Cell proliferation, cell cycle and apoptosis were analyzed by FACS. RESULTS: Gal-8 as a soluble stimulus triggered chemotactic migration of U87 cells across the polycarbonate filter of transwell chambers, almost as intensively as fetal bovine serum. Unexpectedly, Gal-8 also enhanced U87 cell growth. Co-incubation of Gal-8 with lactose, which blocks galectin-glycan interactions, abrogated both effects. Immunoblot showed Gal-8 in conditioned media reflecting its secretion. U87 cells transduced with silencing shRNA in a lentiviral vector expressed and secreted 30-40 % of their normal Gal-8 levels. These cells maintained their migratory capabilities, but decreased their proliferation rate and underwent higher levels of apoptosis, as revealed by flow cytometry analysis of cell cycle, CFSE and activated caspase-3 staining. Proliferation seemed to be more sensitive than migration to Gal-8 expression levels. CONCLUSIONS: Gal-8, either secreted or exogenously enriched in the media, and acting through extracellular glycan interactions, constitutes a strong stimulus of directional migration in glioblastoma U87 cells and for the first time emerges as a factor that promotes proliferation and prevents apoptosis in cancerous cells. These properties could potentially contribute to the exaggerated malignancy of glioblastoma cells.


Subject(s)
Brain Neoplasms/pathology , Galectins/physiology , Glioblastoma/pathology , Animals , Apoptosis/physiology , Brain Neoplasms/genetics , Cattle , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Flow Cytometry/methods , Galectin 1/analysis , Galectin 1/physiology , Galectin 3/analysis , Galectin 3/physiology , Galectins/analysis , Galectins/pharmacology , Glioblastoma/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured
14.
Discov Med ; 21(117): 331-40, 2016 05.
Article in English | MEDLINE | ID: mdl-27355329

ABSTRACT

According to recent studies, mesenchymal stromal cells (MSCs) transplanted via local or tail vein injection can improve healing after anal sphincter injury (ASI) in animal models. However, the transplanted MSCs do not generate skeletal muscle that completely resembles the natural anal sphincter structure. In the present study, we investigated whether bone marrow (BM)-derived MSCs could be induced by Galectin-1 (Gal-1) to differentiate into skeletal muscle and whether the recellularization of an acellular dermal matrix (ADM) with skeletal muscle-differentiated MSCs represents a promising approach to restore ASI in a rat model. BM-MSCs subjected to adenovirus-mediated transfection with Gal-1-GFP (Ad-GFP-Gal-1) displayed increased Gal-1 and desmin expression and differentiated into skeletal muscle cells. MSCs transfected with Ad-GFP-Gal-1 (MSC-Gal-1) were seeded onto an ADM (ADM-MSC-Gal-1) via co-culture, and fusion was observed using a confocal laser scanning microscope. ADM-MSC-Gal-1, ADM-MSC, ADM-MSC-Ad, ADM, or a saline control was applied to a rat ASI model, and injury healing was evaluated via histological examination 6 weeks following treatment. ADM-MSC-Gal-1 treatment promoted significant healing after ASI and improved external anal sphincter contraction curves compared with the other treatments and also led to substantial skeletal muscle regeneration and neovascularization. Our results indicate that repair using ADMs and differentiated MSCs may improve muscle regeneration and restore ASI.


Subject(s)
Anal Canal/injuries , Cell Differentiation/physiology , Galectin 1/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Muscle, Skeletal/physiology , Regeneration/physiology , Wound Healing/physiology , Acellular Dermis , Adenoviridae/genetics , Animals , Cell Line , Disease Models, Animal , Feasibility Studies , Flow Cytometry , Galectin 1/genetics , Genetic Vectors/administration & dosage , Humans , Male , Neovascularization, Physiologic , Rats , Rats, Wistar , Transfection
15.
Cytotherapy ; 18(3): 360-70, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26857229

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) have powerful immunosuppressive activity. This function of MSCs is attributed to plethora of the expressed immunosuppressive factors, such as galectin-1 (Gal-1), a pleiotropic lectin with robust anti-inflammatory effect. Nevertheless, whether Gal-1 renders or contributes to the immunosuppressive effect of MSCs has not been clearly established. Therefore, this question was the focus of a complex study. METHODS: MSCs were isolated from bone marrows of wild-type and Gal-1 knockout mice and their in vitro anti-proliferative and apoptosis-inducing effects on activated T cells were examined. The in vivo immunosuppressive activity was tested in murine models of type I diabetes and delayed-type hypersensitivity. RESULTS: Both Gal-1-expressing and -deficient MSCs inhibited T-cell proliferation. Inhibition of T-cell proliferation by MSCs was mediated by nitric oxide but not PD-L1 or Gal-1. In contrast, MSC-derived Gal-1 triggered apoptosis in activated T cells that were directly coupled to MSCs, representing a low proportion of the T-cell population. Furthermore, absence of Gal-1 in MSCs did not affect their in vivo immunosuppressive effect. CONCLUSIONS: These results serve as evidence that Gal-1 does not play a role in the systemic immunosuppressive effect of MSCs. However, a local contribution of Gal-1 to modulation of T-cell response by direct cell-to-cell interaction cannot be excluded. Notably, this study serves a good model to understand how the specificity of a pleiotropic protein depends on the type and localization of the producing effector cell and its target.


Subject(s)
Cell Communication/genetics , Galectin 1/physiology , Immunologic Factors/physiology , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/genetics , Bone Marrow/metabolism , Cell Proliferation/genetics , Cells, Cultured , Galectin 1/genetics , Immunologic Factors/genetics , Immunosuppressive Agents/metabolism , Lymphocyte Activation/genetics , Male , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
16.
Blood ; 127(15): 1886-95, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26888257

ABSTRACT

Non-Hodgkin lymphoma (NHL) is the most commonly diagnosed hematologic cancer of adults in the United States, with the vast majority of NHLs deriving from malignant B lymphocytes that express cell surface CD20. CD20 immunotherapy (rituximab) is widely used to treat NHL, even though the initial effectiveness of rituximab varies widely among patients and typically wanes over time. The mechanisms through which lymphomas initially resist or gain resistance to immunotherapy are not well established. To address this, a preclinical mouse model system was developed to comprehensively identify lymphoma transcriptomic changes that confer resistance to CD20 immunotherapy. The generation of spontaneous primary and familial lymphomas revealed that sensitivity to CD20 immunotherapy was not regulated by differences in CD20 expression, prior exposure to CD20 immunotherapy, or serial in vivo passage. An unbiased forward exome screen of these primary lymphomas was used to validate the utility of this expansive lymphoma cohort, which revealed that increased lymphoma galectin-1 (Gal-1) expression strongly correlated with resistance to immunotherapy. Genetically induced lymphoma Gal-1 expression ablated antibody-dependent lymphoma phagocytosis in vitro and lymphoma sensitivity to CD20 immunotherapy in vivo. Human NHLs also express elevated Gal-1 compared with nonmalignant lymphocytes, demonstrating the ability of this preclinical model system to identify molecular targets that could be relevant to human therapy. This study therefore established a powerful preclinical model system that permits the comprehensive identification of the dynamic lymphoma molecular network that drives resistance to immunotherapy.


Subject(s)
Antigens, CD20/genetics , Drug Resistance, Neoplasm , Galectin 1/physiology , Immunotherapy/methods , Lymphoma, B-Cell/immunology , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Hemizygote , Humans , Macrophages/cytology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Phagocytosis , Rituximab/therapeutic use
17.
Biol. Res ; 49: 1-10, 2016. ilus, graf
Article in English | LILACS | ID: biblio-950860

ABSTRACT

BACKGROUND: Glioblastoma is one of the most aggressive cancers of the brain. Malignant traits of glioblastoma cells include elevated migration, proliferation and survival capabilities. Galectins are unconventionally secreted glycan-binding proteins that modulate processes of cell adhesion, migration, proliferation and apoptosis by interacting with beta-galactosides of cell surface glycoproteins and the extracellular matrix. Galectin-8 is one of the galectins highly expressed in glioblastoma cells. It has a unique selectivity for terminally sialylated glycans recently found enhanced in these highly malignant cells. A previous study in glioblastoma cell lines reported that Gal-8 coating a plastic surface stimulates two-dimensional motility. Because in other cells Gal-8 arrests proliferation and induces apoptosis, here we extend its study by analyzing all of these processes in a U87 glioblastoma cell mode.l METHODS: We used immunoblot and RT-PCR for Gal-8 expression analysis, recombinant Gal-8 produced in a bacteria system for Gal-8 treatment of the cells, and shRNA in lentivirus transduction for Gal-8 silencing. Cell migration as assessed in transwell filters. Cell proliferation, cell cycle and apoptosis were analyzed by FACS. RESULTS: Gal-8 as a soluble stimulus triggered chemotactic migration of U87 cells across the polycarbonate filter of transwell chambers, almost as intensively as fetal bovine serum. Unexpectedly, Gal-8 also enhanced U87 cell growth. Co-incubation of Gal-8 with lactose, which blocks galectin-glycan interactions, abrogated both effects. Immunoblot showed Gal-8 in conditioned media reflecting its secretion. U87 cells transduced with silencing shRNA in a lentiviral vector expressed and secreted 30-40 % of their normal Gal-8 levels. These cells maintained their migratory capabilities, but decreased their proliferation rate and underwent higher levels of apoptosis, as revealed by flow cytometry analysis of cell cycle, CFSE and activated caspase-3 staining. Proliferation seemed to be more sensitive than migration to Gal-8 expression levels. CONCLUSIONS: Gal-8, either secreted or exogenously enriched in the media, and acting through extracellular glycan interactions, constitutes a strong stimulus of directional migration in glioblastoma U87 cells and for the first time emerges as a factor that promotes proliferation and prevents apoptosis in cancerous cells. These properties could potentially contribute to the exaggerated malignancy of glioblastoma cells.


Subject(s)
Humans , Animals , Cattle , Brain Neoplasms/pathology , Glioblastoma/pathology , Galectins/physiology , Time Factors , Brain Neoplasms/genetics , Tumor Cells, Cultured , Cell Movement/physiology , Apoptosis/physiology , Glioblastoma/genetics , Reverse Transcriptase Polymerase Chain Reaction , Galectins/analysis , Galectins/pharmacology , Galectin 1/analysis , Galectin 1/physiology , Galectin 3/analysis , Galectin 3/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Flow Cytometry/methods
18.
Cancer Biother Radiopharm ; 30(8): 336-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26348206

ABSTRACT

Galectin-1 is a member of carbohydrate-binding proteins and plays critical roles in tumor growth and progression. It has been reported that galectin-1 is upregulated in human hepatocellular carcinoma (HCC) and facilitates HCC cell migration and invasion. In this study, the authors aimed to explore the effects of the knockdown of galectin-1 on HCC cell survival and sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Human HCC cells were transfected with galectin-1-targeting small interfering RNA (siRNA) with or without 100 ng/mL TRAIL treatment and tested for apoptosis and gene expression changes. Cotransfection of Bcl-2- and survivin-expressing plasmids with galectin-1 siRNA was done, before TRAIL exposure, cell viability, and apoptosis were assessed. The authors found that siRNA-mediated downregulation of galectin-1 caused apoptosis in HCC cells, which was coupled with reduced Bcl-2 and survivin and increased Bax expression. Overexpression of Bcl-2 and survivin significantly blocked galectin-1 silencing-induced apoptosis of HCC cells. Knockdown of galectin-1 significantly enhanced TRAIL cytotoxicity against HCC cells, as determined by the MTT assay. Moreover, galectin-1 downregulation significantly induced apoptosis in TRAIL-treated HCC cells. Such effects were almost completely counteracted by the enforced expression of Bcl-2 and survivin. Taken together, these data first show that galectin-1 downregulation induces apoptosis in and augments TRAIL cytotoxicity to HCC cells largely through regulation of Bcl-2 and survivin expression. These findings provide a rationale for preclinical and clinical evaluation of targeting galectin-1 for improving TRAIL-based therapy against HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Galectin 1/physiology , Liver Neoplasms/pathology , Neoplasm Proteins/physiology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Survival , Down-Regulation , Drug Resistance, Neoplasm , Galectin 1/antagonists & inhibitors , Galectin 1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Genes, Reporter , Genetic Vectors , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/physiology , RNA Interference , Recombinant Proteins/pharmacology , Survivin , Transfection
19.
Mol Reprod Dev ; 82(10): 765-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26096842

ABSTRACT

Our previous findings showed that galectin-1 (LGALS1) plays an important role in the in vitro invasion of normal human trophoblast cells. In the present study, choriocarcinoma JAr cells were found to express LGALS1, -2, -3, -8, -10, and -13 mRNA and at least LGALS1, -3, and -8 protein, as determined by reverse-transcriptase PCR and Western blot, respectively. The galectin mRNA signature of JAr cells thus differed from that of normal first-trimester extravillous trophoblasts. A Matrigel migration assay was also used to investigate and confirm the relevance and effect of LGALS1 on the invasive potential of JAr cells, as observed in other trophoblast models. This modulation in behavior was achieved by specific lectin-glycan binding.


Subject(s)
Choriocarcinoma/metabolism , Galectin 1/biosynthesis , Neoplasm Invasiveness , Placentation , Trophoblasts/metabolism , Cell Line, Tumor , Choriocarcinoma/pathology , Female , Galectin 1/genetics , Galectin 1/physiology , Gene Expression Profiling , Humans , Pregnancy
20.
PLoS One ; 9(9): e107933, 2014.
Article in English | MEDLINE | ID: mdl-25243744

ABSTRACT

Despite their abundance at gastrointestinal sites, little is known about the role of galectins in gut immune responses. We have therefore investigated the Citrobacter rodentium model of colonic infection and inflammation in Galectin-1 or Galectin-3 null mice. Gal-3 null mice showed a slight delay in colonisation after inoculation with C. rodentium and a slight delay in resolution of infection, associated with delayed T cell, macrophage and dendritic cell infiltration into the gut mucosa. However, Gal-1 null mice also demonstrated reduced T cell and macrophage responses to infection. Despite the reduced T cell and macrophage response in Gal-1 null mice, there was no effect on C. rodentium infection kinetics and pathology. Overall, Gal-1 and Gal-3 play only a minor role in immunity to a gut bacterial pathogen.


Subject(s)
Citrobacter rodentium/isolation & purification , Enterobacteriaceae Infections/immunology , Galectin 1/physiology , Galectin 3/physiology , Immunity, Mucosal/physiology , Animals , Galectin 1/genetics , Galectin 3/genetics , Interleukin-6/metabolism , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...