Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 30(4): 906-921, 2023 04.
Article in English | MEDLINE | ID: mdl-36693903

ABSTRACT

Non-melanoma skin cancer (NMSC) has risen dramatically as a result of chronic exposure to sunlight ultraviolet (UV) radiation, climatic changes and clinical conditions associated with immunosuppression. In spite of considerable progress, our understanding of the mechanisms that control NMSC development and their associated molecular and immunological landscapes is still limited. Here we demonstrated a critical role for galectin-7 (Gal-7), a ß-galactoside-binding protein preferentially expressed in skin tissue, during NMSC development. Transgenic mice (Tg46) overexpressing Gal-7 in keratinocytes showed higher number of papillomas compared to WT mice or mice lacking Gal-7 (Lgals7-/-) when subjected to a skin carcinogenesis protocol, in which tumor initiator 7,12-dimethylbenz[a]anthracene (DMBA) and tumor promoter 12-O-tetradecanoyl-phorbol-13-acetate (TPA) were sequentially administered. RNAseq analysis of Tg46 tumor lesions revealed a unique profile compatible with cells of the myelomonocytic lineage infiltrating these tumors, an effect that was substantiated by a higher number of CD11b+Gr1+ cells in tumor-draining lymph nodes. Heightened c-Met activation and Cxcl-1 expression in Tg46 lesions suggested a contribution of this pathway to the recruitment of these cells. Remarkably, Gal-7 bound to the surface of CD11b+Ly6ChiLy6Glo monocytic myeloid cells and enhanced their immunosuppressive activity, as evidenced by increased IL-10 and TGF-ß1 secretion, and higher T-cell inhibitory activity. In vivo, carcinogen-treated Lgals7-/- animals adoptively transferred with Gal-7-conditioned monocytic myeloid cells developed higher number of papillomas, whereas depletion of these cells in Tg46-treated mice led to reduction in the number of tumors. Finally, human NMSC biopsies showed increased LGALS7 mRNA and Gal-7 protein expression and displayed transcriptional profiles associated with myeloid programs, accompanied by elevated CXCL1 expression and c-Met activation. Thus, Gal-7 emerges as a critical mediator of skin carcinogenesis and a potential therapeutic target in human NMSC.


Subject(s)
Papilloma , Skin Neoplasms , Mice , Animals , Humans , Carcinogens , Skin Neoplasms/pathology , Papilloma/pathology , Carcinogenesis/genetics , Mice, Transgenic , Galectins/genetics , Skin/metabolism , Immunity, Innate
2.
Methods Mol Biol ; 2442: 635-653, 2022.
Article in English | MEDLINE | ID: mdl-35320550

ABSTRACT

Development of an aberrant vascular network is a hallmark of the multistep pathological process of tumor growth and metastasis. In response to hypoxia, several pro-angiogenic factors are synthesized to support vascularization programs required for cancer progression. Emerging data indicate the involvement of glycans and glycan-binding proteins as critical regulators of vascular circuits in health and disease. Galectins may be regulated by hypoxic conditions and control angiogenesis in different physiopathological settings. These ß-galactoside-binding proteins may promote sprouting angiogenesis by interacting with different glycosylated receptors and triggering distinct signaling pathways. Understanding the role of galectins in tumor neovascularization will contribute to the design of novel anti-angiogenic therapies aimed at complementing current anti-cancer modalities and overcoming resistance to these treatments. Here we describe selected strategies and methods used to study the role of hypoxia-regulated galectins in the regulation of blood vessel formation.


Subject(s)
Galectins , Hypoxia , Neoplasms , Neovascularization, Pathologic , Galectins/metabolism , Humans , Hypoxia/physiopathology , Neoplasms/blood supply , Neovascularization, Pathologic/physiopathology , Signal Transduction
4.
Angiogenesis ; 24(1): 1-5, 2021 02.
Article in English | MEDLINE | ID: mdl-33006019

ABSTRACT

Galectins, a family of highly conserved ß-galactoside-binding proteins, control tumor progression by modulating different hallmarks of cancer. Galectin-1 (Gal-1), a proto-type member of this family, plays essential roles in tumor angiogenesis and immunosuppression by cross-linking glycosylated receptors on the surface of endothelial and immune cells. Targeted disruption of Gal-1 suppresses tumor growth by counteracting aberrant angiogenesis and reinforcing antitumor immunity in several experimental settings. Given the multiple therapeutic benefits associated with Gal-1 blockade, several Gal-1 inhibitors, including glycan-based competitors, antagonistic peptides, aptamers and neutralizing monoclonal antibodies, have been designed and evaluated in pre-clinical tumor models. Here we report the biochemical and functional characterization of a newly developed neutralizing anti-human Gal-1 monoclonal antibody (Gal-1-mAb3), which specifically recognizes a unique epitope in Gal-1 protein and exerts both angioregulatory and immunomodulatory activities. Blockade of Gal-1 function using Gal-1-mAb3, might be relevant not only in cancer but also in other pathologic conditions characterized by aberrant angiogenesis and uncontrolled immunosuppression.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Galectin 1/immunology , Immunologic Factors/pharmacology , Neovascularization, Physiologic , Animals , Biophysical Phenomena , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects
5.
J Exp Med ; 217(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32196081

ABSTRACT

The activity and survival of retinal photoreceptors depend on support functions performed by the retinal pigment epithelium (RPE) and on oxygen and nutrients delivered by blood vessels in the underlying choroid. By combining single-cell and bulk RNA sequencing, we categorized mouse RPE/choroid cell types and characterized the tissue-specific transcriptomic features of choroidal endothelial cells. We found that choroidal endothelium adjacent to the RPE expresses high levels of Indian Hedgehog and identified its downstream target as stromal GLI1+ mesenchymal stem cell-like cells. In vivo genetic impairment of Hedgehog signaling induced significant loss of choroidal mast cells, as well as an altered inflammatory response and exacerbated visual function defects after retinal damage. Our studies reveal the cellular and molecular landscape of adult RPE/choroid and uncover a Hedgehog-regulated choroidal immunomodulatory signaling circuit. These results open new avenues for the study and treatment of retinal vascular diseases and choroid-related inflammatory blinding disorders.


Subject(s)
Choroid/immunology , Choroid/pathology , Endothelium/immunology , Immunomodulation , Single-Cell Analysis , Animals , Cell Proliferation , Endothelial Cells/metabolism , Gene Expression Regulation , Hedgehog Proteins/metabolism , Inflammation/genetics , Mast Cells/metabolism , Melanocytes/metabolism , Melanocytes/pathology , Mice, Inbred C57BL , Organ Specificity , Retinal Pigment Epithelium/metabolism , Signal Transduction , Transcription, Genetic , Zinc Finger Protein GLI1/metabolism
6.
Nature ; 562(7727): 423-428, 2018 10.
Article in English | MEDLINE | ID: mdl-30305738

ABSTRACT

Tumours evade immune control by creating hostile microenvironments that perturb T cell metabolism and effector function1-4. However, it remains unclear how intra-tumoral T cells integrate and interpret metabolic stress signals. Here we report that ovarian cancer-an aggressive malignancy that is refractory to standard treatments and current immunotherapies5-8-induces endoplasmic reticulum stress and activates the IRE1α-XBP1 arm of the unfolded protein response9,10 in T cells to control their mitochondrial respiration and anti-tumour function. In T cells isolated from specimens collected from patients with ovarian cancer, upregulation of XBP1 was associated with decreased infiltration of T cells into tumours and with reduced IFNG mRNA expression. Malignant ascites fluid obtained from patients with ovarian cancer inhibited glucose uptake and caused N-linked protein glycosylation defects in T cells, which triggered IRE1α-XBP1 activation that suppressed mitochondrial activity and IFNγ production. Mechanistically, induction of XBP1 regulated the abundance of glutamine carriers and thus limited the influx of glutamine that is necessary to sustain mitochondrial respiration in T cells under glucose-deprived conditions. Restoring N-linked protein glycosylation, abrogating IRE1α-XBP1 activation or enforcing expression of glutamine transporters enhanced mitochondrial respiration in human T cells exposed to ovarian cancer ascites. XBP1-deficient T cells in the metastatic ovarian cancer milieu exhibited global transcriptional reprogramming and improved effector capacity. Accordingly, mice that bear ovarian cancer and lack XBP1 selectively in T cells demonstrate superior anti-tumour immunity, delayed malignant progression and increased overall survival. Controlling endoplasmic reticulum stress or targeting IRE1α-XBP1 signalling may help to restore the metabolic fitness and anti-tumour capacity of T cells in cancer hosts.


Subject(s)
Endoribonucleases/metabolism , Mitochondria/metabolism , Ovarian Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , X-Box Binding Protein 1/metabolism , Amino Acid Transport Systems, Basic , Animals , Ascites/metabolism , Cell Respiration , Disease Progression , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glutamine/metabolism , Glycosylation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Signal Transduction , Survival Rate , T-Lymphocytes/metabolism , Tumor Escape/immunology , Unfolded Protein Response , X-Box Binding Protein 1/biosynthesis , X-Box Binding Protein 1/deficiency
7.
Handb Exp Pharmacol ; 249: 31-61, 2018.
Article in English | MEDLINE | ID: mdl-28405776

ABSTRACT

In contrast to mechanisms taking place during resistance to chemotherapies or other targeted therapies, compensatory adaptation to angiogenesis blockade does not imply a mutational alteration of genes encoding drug targets or multidrug resistance mechanisms but instead involves intrinsic or acquired activation of compensatory angiogenic pathways. In this article we highlight hypoxia-regulated and immune-mediated mechanisms that converge in endothelial cell programs and preserve angiogenesis in settings of vascular endothelial growth factor (VEGF) blockade. These mechanisms involve mobilization of myeloid cell populations and activation of cytokine- and chemokine-driven circuits operating during intrinsic and acquired resistance to anti-angiogenic therapies. Particularly, we focus on findings underscoring a role for galectins and glycosylated ligands in promoting resistance to anti-VEGF therapies and discuss possible strategies to overcome or attenuate this compensatory pathway. Finally, we highlight emerging evidence demonstrating the interplay between immunosuppressive and pro-angiogenic programs in the tumor microenvironment (TME) and discuss emerging combinatorial anticancer strategies aimed at simultaneously potentiating antitumor immune responses and counteracting aberrant angiogenesis.


Subject(s)
Hypoxia , Neoplasms/drug therapy , Neoplasms/immunology , Neovascularization, Pathologic/drug therapy , Chemokines , Cytokines , Drug Resistance, Neoplasm , Galectins , Humans , Tumor Microenvironment , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Cell Death Dis ; 8(12): e3176, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215607

ABSTRACT

Secretory granules released by cytotoxic T lymphocytes (CTLs) are powerful weapons against intracellular microbes and tumor cells. Despite significant progress, there is still limited information on the molecular mechanisms implicated in target-driven degranulation, effector cell survival and composition and structure of the lytic granules. Here, using a proteomic approach we identified a panel of putative cytotoxic granule proteins, including some already known granule constituents and novel proteins that contribute to regulate the CTL lytic machinery. Particularly, we identified galectin-1 (Gal1), an endogenous immune regulatory lectin, as an integral component of the secretory granule machinery and unveil the unexpected function of this lectin in regulating CTL killing activity. Mechanistic studies revealed the ability of Gal1 to control the non-secretory lytic pathway by influencing Fas-Fas ligand interactions. This study offers new insights on the composition of the cytotoxic granule machinery, highlighting the dynamic cross talk between secretory and non-secretory pathways in controlling CTL lytic function.


Subject(s)
Cell Degranulation/immunology , Cytotoxicity, Immunologic , Fas Ligand Protein/genetics , Galectin 1/genetics , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/genetics , Animals , Cell Proliferation , Fas Ligand Protein/immunology , Galectin 1/immunology , Gene Expression Profiling , Gene Expression Regulation , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteomics , Secretory Vesicles/chemistry , Secretory Vesicles/immunology , Secretory Vesicles/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/cytology , fas Receptor/immunology
9.
Trends Biochem Sci ; 42(4): 255-273, 2017 04.
Article in English | MEDLINE | ID: mdl-27986367

ABSTRACT

The vast range and complexity of glycan structures and their dynamic variations in health and disease have presented formidable challenges toward understanding the biological significance of these molecules. Despite these limitations, compelling evidence highlights a major role for galectins, a family of soluble glycan-binding proteins, as endogenous decoders that translate glycan-containing information into a broad spectrum of cellular responses by modulating receptor clustering, reorganization, endocytosis, and signaling. Here, we underscore pioneer findings and recent advances in understanding the biology of galectin-glycan interactions in myeloid, lymphoid, and endothelial compartments, highlighting important pathways by which these multivalent complexes control immune and vascular programs. Implementation of novel glycoanalytical approaches, as well as the use of genetically engineered cell and organism models, have allowed glycans and galectins to be explored across a range of cellular processes.


Subject(s)
Endothelium/metabolism , Immune System/metabolism , Polysaccharides/immunology , Polysaccharides/metabolism , Signal Transduction , Galectins/metabolism , Humans , Polysaccharides/chemistry
10.
Oncotarget ; 7(22): 31738-54, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27166189

ABSTRACT

Galectin-1 (Gal1) is a known immune/inflammatory regulator which acts both extracellularly and intracellularly, modulating innate and adaptive immune responses. Here, we explored the role of Gal1 in liver regeneration using 70% partial hepatectomy (PHx) of C57BL/6 wild type and Gal1-knockout (Gal1-KO, Lgals1-/-) mice. Gene or protein expression, in liver samples collected at time intervals from 2 to 168 hours post-operation, was tested by either RT-PCR or by immunoblotting and immunohistochemistry, respectively. We demonstrated that Gal1 transcript and protein expression was induced in the liver tissue of wild type mice upon PHx. Liver regeneration following PHx was significantly delayed in the Gal1-KO compared to the control liver. This delay was accompanied by a decreased Akt phosphorylation, and accumulation of the hepatocyte nuclear p21 protein in the Gal1-KO versus control livers at 24 and 48 hours following PHx. Transcripts of several known regulators of inflammation, cell cycle and cell signaling, including some known PHx-induced genes, were aberrantly expressed (mainly down-regulated) in Gal1-KO compared to control livers at 2, 6 and 24 hours post-PHx. Transient steatosis, which is imperative for liver regeneration following PHx, was significantly delayed and decreased in the Gal1-KO compared to the control liver and was accompanied by a significantly decreased expression in the mutant liver of several genes encoding lipid metabolism regulators. Our results demonstrate that Gal1 protein is essential for efficient liver regeneration following PHx through the regulation of liver inflammation, hepatic cell proliferation, and the control of lipid storage in the regenerating liver.


Subject(s)
Cell Proliferation , Galectin 1/metabolism , Hepatectomy , Liver Regeneration , Liver/surgery , Animals , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Galectin 1/deficiency , Galectin 1/genetics , Genotype , Hepatitis/genetics , Hepatitis/metabolism , Hepatitis/pathology , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors
11.
PLoS Negl Trop Dis ; 9(10): e0004148, 2015.
Article in English | MEDLINE | ID: mdl-26451839

ABSTRACT

BACKGROUND: Chronic Chagas cardiomyopathy caused by Trypanosoma cruzi is the result of a pathologic process starting during the acute phase of parasite infection. Among different factors, the specific recognition of glycan structures by glycan-binding proteins from the parasite or from the mammalian host cells may play a critical role in the evolution of the infection. METHODOLOGY AND PRINCIPAL FINDINGS: Here we investigated the contribution of galectin-1 (Gal-1), an endogenous glycan-binding protein abundantly expressed in human and mouse heart, to the pathophysiology of T. cruzi infection, particularly in the context of cardiac pathology. We found that exposure of HL-1 cardiac cells to Gal-1 reduced the percentage of infection by two different T. cruzi strains, Tulahuén (TcVI) and Brazil (TcI). In addition, Gal-1 prevented exposure of phosphatidylserine and early events in the apoptotic program by parasite infection on HL-1 cells. These effects were not mediated by direct interaction with the parasite surface, suggesting that Gal-1 may act through binding to host cells. Moreover, we also observed that T. cruzi infection altered the glycophenotype of cardiac cells, reducing binding of exogenous Gal-1 to the cell surface. Consistent with these data, Gal-1 deficient (Lgals1-/-) mice showed increased parasitemia, reduced signs of inflammation in heart and skeletal muscle tissues, and lower survival rates as compared to wild-type (WT) mice in response to intraperitoneal infection with T. cruzi Tulahuén strain. CONCLUSION/SIGNIFICANCE: Our results indicate that Gal-1 modulates T. cruzi infection of cardiac cells, highlighting the relevance of galectins and their ligands as regulators of host-parasite interactions.


Subject(s)
Chagas Disease/immunology , Chagas Disease/pathology , Galectin 1/metabolism , Host-Parasite Interactions , Myocytes, Cardiac/physiology , Myocytes, Cardiac/parasitology , Trypanosoma cruzi/immunology , Adult , Aged , Animals , Brazil , Cells, Cultured , Chagas Disease/parasitology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Parasitemia , Survival Analysis
12.
J Immunol ; 195(7): 3311-24, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324777

ABSTRACT

Galectin-1 (Gal-1), an endogenous glycan-binding protein, is widely distributed at sites of inflammation and microbial invasion. Despite considerable progress regarding the immunoregulatory activity of this lectin, the role of endogenous Gal-1 during acute parasite infections is uncertain. In this study, we show that Gal-1 functions as a negative regulator to limit host-protective immunity following intradermal infection with Trypanosoma cruzi. Concomitant with the upregulation of immune inhibitory mediators, including IL-10, TGF-ß1, IDO, and programmed death ligand 2, T. cruzi infection induced an early increase of Gal-1 expression in vivo. Compared to their wild-type (WT) counterpart, Gal-1-deficient (Lgals1(-/-)) mice exhibited reduced mortality and lower parasite load in muscle tissue. Resistance of Lgals1(-/-) mice to T. cruzi infection was associated with a failure in the activation of Gal-1-driven tolerogenic circuits, otherwise orchestrated by WT dendritic cells, leading to secondary dysfunction in the induction of CD4(+)CD25(+)Foxp3(+) regulatory T cells. This effect was accompanied by an increased number of CD8(+) T cells and higher frequency of IFN-γ-producing CD4(+) T cells in muscle tissues and draining lymph nodes as well as reduced parasite burden in heart and hindlimb skeletal muscle. Moreover, dendritic cells lacking Gal-1 interrupted the Gal-1-mediated tolerogenic circuit and reinforced T cell-dependent anti-parasite immunity when adoptively transferred into WT mice. Thus, endogenous Gal-1 may influence T. cruzi infection by fueling tolerogenic circuits that hinder anti-parasite immunity.


Subject(s)
Chagas Disease/immunology , Dendritic Cells/immunology , Galectin 1/genetics , T-Lymphocytes, Regulatory/immunology , Trypanosoma cruzi/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/mortality , Chagas Disease/parasitology , Galectin 1/biosynthesis , Galectin 1/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Knockout , Parasite Load , Programmed Cell Death 1 Ligand 2 Protein/biosynthesis , Transforming Growth Factor beta1/biosynthesis
13.
Stem Cells ; 33(3): 646-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25407338

ABSTRACT

Many solid tumors contain a subpopulation of cells with stem characteristics and these are known as cancer stem cells (CSCs) or tumor-initiating cells (TICs). These cells drive tumor growth and appear to be regulated by molecular pathway different from other cells in the tumor bulk. Here, we set out to determine whether elements of the PI3K-AKT pathway are necessary to maintain the CSC-like phenotype in breast tumor cells and for these cells to survive, bearing in mind that the identification of such elements is likely to be relevant to define future therapeutic targets. Our results demonstrate a close relationship between the maintenance of the CSC-like phenotype and the survival of these TICs. Inhibiting PI3K activity, or eliminating AKT activity, mostly that of the AKT1 isoform, produces a clear drop in TICs survival, and a reduction in the generation and growth of CD44(High) /CD24(Low) mammospheres. Surprisingly, the apoptosis of these TICs that is triggered by AKT1 deficiency is also associated with a loss of the stem cell/mesenchymal phenotype and a recovery of epithelial-like markers. Finally, we define downstream effectors that are responsible for controlling the CSC-phenotype, such as FoxO-Bim, and the death of these cells in the absence of AKT1. In summary, these data closely link the maintenance of the stem cell-like phenotype and the survival of these cells to the AKT-FoxO-Bim pathway.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Proliferation/physiology , Female , Forkhead Box Protein O1 , Humans , MCF-7 Cells , Phenotype , Signal Transduction
14.
Methods Mol Biol ; 1207: 249-68, 2015.
Article in English | MEDLINE | ID: mdl-25253145

ABSTRACT

During the past decade, a better understanding of the cellular and molecular mechanisms underlying tumor immunity has provided the appropriate framework for the development of therapeutic strategies for cancer immunotherapy. Under this complex scenario, galectins have emerged as promising molecular targets for cancer therapy responsible of creating immunosuppressive microenvironments at sites of tumor growth and metastasis. Galectins, expressed in tumor, stromal, and endothelial cells, contribute to thwart the development of immune responses by favoring the expansion of T regulatory cells and contributing to their immunosuppressive activity, driving the differentiation of tolerogenic dendritic cells, limiting T cell viability, and maintaining T cell anergy. The emerging data promise a future scenario in which the selective blockade of individual members of the galectin family, either alone or in combination with other therapeutic regimens, will contribute to halt tumor progression by counteracting tumor-immune escape. Here we describe a selection of methods used to investigate the role of galectin-1 in tumor-immune escape.


Subject(s)
Galectins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Adoptive Transfer , Animals , Antigens, Neoplasm/immunology , Blotting, Western , Bone Marrow Cells/cytology , CD3 Complex/metabolism , Cell Proliferation , Cell Separation , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Galectin 1/metabolism , Gene Silencing , Genetic Vectors/genetics , Humans , Interleukin-27/metabolism , Lentivirus/genetics , Lymph Nodes/immunology , Mice , Neoplasms/pathology , Phosphorylation , STAT3 Transcription Factor/metabolism , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transduction, Genetic , Tumor Microenvironment
15.
Methods Mol Biol ; 1207: 293-304, 2015.
Article in English | MEDLINE | ID: mdl-25253148

ABSTRACT

Formation of an aberrant and heterogeneous vascular network is a key pathological event in the multistep process of tumor growth and metastasis. Pro-angiogenic factors are synthesized and released from tumor, stromal, endothelial, and myeloid cells in response to hypoxic and immunosuppressive microenvironments which are commonly found during cancer progression. Emerging data indicate key roles for galectins, particularly galectin-1, -3, -8, and -9 in the regulation of angiogenesis in different pathophysiologic settings. Each galectin interacts with a preferred set of glycosylated receptors, triggers different signaling pathway, and promotes sprouting angiogenesis through different mechanisms. Understanding the role of galectins in tumor neovascularization will contribute to the design of novel anti-angiogenic therapies aimed at complementing current clinical approaches. Here we describe selected strategies and methods used to study the galectin-1 regulation by hypoxia and its role in blood vessel formation.


Subject(s)
Galectin 1/metabolism , Neovascularization, Pathologic/metabolism , Blotting, Western , Cell Hypoxia , Cell Movement , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Solubility , Tumor Microenvironment , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
16.
Glycobiology ; 24(12): 1283-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25117007

ABSTRACT

Abnormal glycosylation is a typical hallmark of the transition from healthy to neoplastic tissues. Although the importance of glycans and glycan-binding proteins in cancer-related processes such as tumor cell adhesion, migration, metastasis and immune escape has been largely appreciated, our awareness of the impact of lectin-glycan recognition in tumor vascularization is relatively new. Regulated glycosylation can influence vascular biology by controlling trafficking, endocytosis and signaling of endothelial cell (EC) receptors including vascular endothelial growth factor receptors, platelet EC adhesion molecule, Notch and integrins. In addition, glycans may control angiogenesis by regulating migration of endothelial tip cells and influencing EC survival and vascular permeability. Recent evidence indicated that changes in the EC surface glycome may also serve "on-and-off" switches that control galectin binding to signaling receptors by displaying or masking-specific glycan epitopes. These glycosylation-dependent lectin-receptor interactions can link tumor hypoxia to EC signaling and control tumor sensitivity to anti-angiogenic treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Hypoxia/metabolism , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Polysaccharides/metabolism , Endothelial Cells/metabolism , Glycosylation , Humans , Signal Transduction
17.
Cell ; 156(4): 744-58, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24529377

ABSTRACT

The clinical benefit conferred by vascular endothelial growth factors (VEGF)-targeted therapies is variable, and tumors from treated patients eventually reinitiate growth. Here, we identify a glycosylation-dependent pathway that compensates for the absence of cognate ligand and preserves angiogenesis in response to VEGF blockade. Remodeling of the endothelial cell (EC) surface glycome selectively regulated binding of galectin-1 (Gal1), which upon recognition of complex N-glycans on VEGFR2, activated VEGF-like signaling. Vessels within anti-VEGF-sensitive tumors exhibited high levels of α2-6-linked sialic acid, which prevented Gal1 binding. In contrast, anti-VEGF refractory tumors secreted increased Gal1 and their associated vasculature displayed glycosylation patterns that facilitated Gal1-EC interactions. Interruption of ß1-6GlcNAc branching in ECs or silencing of tumor-derived Gal1 converted refractory into anti-VEGF-sensitive tumors, whereas elimination of α2-6-linked sialic acid conferred resistance to anti-VEGF. Disruption of the Gal1-N-glycan axis promoted vascular remodeling, immune cell influx and tumor growth inhibition. Thus, targeting glycosylation-dependent lectin-receptor interactions may increase the efficacy of anti-VEGF treatment.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic , Vascular Endothelial Growth Factors/antagonists & inhibitors , Animals , Endothelial Cells/metabolism , Galectin 1/genetics , Galectin 1/metabolism , Glycosylation , Humans , Hypoxia , Mice , Receptors, Mitogen/metabolism
18.
Am J Pathol ; 182(1): 29-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142379

ABSTRACT

Galectin-1 (Gal-1), an evolutionarily conserved ß-galactoside-binding lectin, plays essential roles in the control of inflammation and neovascularization. Although identified as a major component of the contractile apparatus of cardiomyocytes, the potential role of Gal-1 in modulating heart pathophysiology is uncertain. Here, we aimed to characterize Gal-1 expression and function in the infarcted heart. Expression of Gal-1 was substantially increased in the mouse heart 7 days after acute myocardial infarction (AMI) and in hearts from patients with end-stage chronic heart failure. This lectin was localized mainly in cardiomyocytes and inflammatory infiltrates in peri-infarct areas, but not in remote areas. Both simulated hypoxia and proinflammatory cytokines selectively up-regulated Gal-1 expression in mouse cardiomyocytes, whereas anti-inflammatory cytokines inhibited expression of this lectin or had no considerable effect. Compared with their wild-type counterpart, Gal-1-deficient (Lgals1(-/-)) mice showed enhanced cardiac inflammation, characterized by increased numbers of macrophages, natural killer cells, and total T cells, but reduced frequency of regulatory T cells, leading to impaired cardiac function at baseline and impaired ventricular remodeling 7 days after nonreperfused AMI. Treatment of mice with recombinant Gal-1 attenuated cardiac damage in reperfused AMI. Taken together, our results indicate a protective role for Gal-1 in normal cardiac homeostasis and postinfarction remodeling by preventing cardiac inflammation. Thus, Gal-1 treatment represents a potential novel strategy to attenuate heart failure in AMI.


Subject(s)
Galectin 1/physiology , Myocardial Infarction/physiopathology , Myocarditis/metabolism , Ventricular Remodeling/physiology , Adult , Aged , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Hypoxia/physiology , Cells, Cultured , Cytokines/pharmacology , Female , Galectin 1/biosynthesis , Galectin 1/pharmacology , Galectin 1/therapeutic use , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Inflammation Mediators/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocarditis/etiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Recombinant Proteins/therapeutic use , Up-Regulation/drug effects , Up-Regulation/physiology , Ventricular Function, Left/physiology , Young Adult
19.
Cancer Res ; 73(3): 1107-17, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23204230

ABSTRACT

Galectin-1 (Gal1), an evolutionarily conserved glycan-binding protein, contributes to the creation of an immunosuppressed microenvironment at sites of tumor growth. In spite of considerable progress in elucidating its role in tumor-immune escape, the mechanisms underlying the inhibitory functions of Gal1 remain obscure. Here, we investigated the contribution of tumor Gal1 to tumor growth, metastasis, and immunosuppression in breast cancer. We found that the frequency of Gal1(+) cells in human breast cancer biopsies correlated positively with tumor grade, while specimens from patients with benign hyperplasia showed negative or limited Gal1 staining. To examine the pathophysiologic relevance of Gal1 in breast cancer, we used the metastatic mouse mammary tumor 4T1, which expresses and secretes substantial amounts of Gal1. Silencing Gal1 expression in this model induced a marked reduction in both tumor growth and the number of lung metastases. This effect was abrogated when mice were inoculated with wild-type 4T1 tumor cells in their contralateral flank, suggesting involvement of a systemic modulation of the immune response. Gal1 attenuation in 4T1 cells also reduced the frequency of CD4(+)CD25(+) Foxp3(+) regulatory T (T(reg)) cells within the tumor, draining lymph nodes, spleen, and lung metastases. Further, it abrogated the immunosuppressive function of T(reg) cells and selectively lowered the expression of the T-cell regulatory molecule LAT (linker for activation of T cells) on these cells, disarming their suppressive activity. Taken together, our results offer a preclinical proof of concept that therapeutic targeting of Gal1 can overcome breast cancer-associated immunosuppression and can prevent metastatic disease.


Subject(s)
Breast Neoplasms/pathology , Galectin 1/physiology , Immune Tolerance , Animals , Breast Neoplasms/immunology , Female , Galectin 1/antagonists & inhibitors , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology
20.
J Exp Med ; 209(11): 1985-2000, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23027923

ABSTRACT

Kaposi's sarcoma (KS), a multifocal vascular neoplasm linked to human herpesvirus-8 (HHV-8/KS-associated herpesvirus [KSHV]) infection, is the most common AIDS-associated malignancy. Clinical management of KS has proven to be challenging because of its prevalence in immunosuppressed patients and its unique vascular and inflammatory nature that is sustained by viral and host-derived paracrine-acting factors primarily released under hypoxic conditions. We show that interactions between the regulatory lectin galectin-1 (Gal-1) and specific target N-glycans link tumor hypoxia to neovascularization as part of the pathogenesis of KS. Expression of Gal-1 is found to be a hallmark of human KS but not other vascular pathologies and is directly induced by both KSHV and hypoxia. Interestingly, hypoxia induced Gal-1 through mechanisms that are independent of hypoxia-inducible factor (HIF) 1α and HIF-2α but involved reactive oxygen species-dependent activation of the transcription factor nuclear factor κB. Targeted disruption of Gal-1-N-glycan interactions eliminated hypoxia-driven angiogenesis and suppressed tumorigenesis in vivo. Therapeutic administration of a Gal-1-specific neutralizing mAb attenuated abnormal angiogenesis and promoted tumor regression in mice bearing established KS tumors. Given the active search for HIF-independent mechanisms that serve to couple tumor hypoxia to pathological angiogenesis, our findings provide novel opportunities not only for treating KS patients but also for understanding and managing a variety of solid tumors.


Subject(s)
Galectin 1/metabolism , Neovascularization, Pathologic/metabolism , Polysaccharides/metabolism , Sarcoma, Kaposi/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Galectin 1/genetics , Galectin 1/immunology , Gene Expression Regulation, Neoplastic , HEK293 Cells , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Humans , Hypoxia , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Kaposi/drug therapy , Sarcoma, Kaposi/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...