1
|
Torres NI, Manselle Cocco MN, Perrotta RM, Mahmoud YD, Salatino M, Mariño KV, Rabinovich GA. A single-step, rapid, and versatile method for simultaneous detection of cell surface glycan profiles using fluorochrome-conjugated lectins. Glycobiology 2023; 33:855-860. [PMID: 37584473 DOI: 10.1093/glycob/cwad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023] Open
Abstract
Cell surface glycans play essential roles in diverse physiological and pathological processes and their assessment has important implications in biomedicine and biotechnology. Here we present a rapid, versatile, and single-step multicolor flow cytometry method for evaluation of cell surface glycan signatures using a panel of selected fluorochrome-conjugated lectins. This procedure allows simultaneous detection of cell surface glycans with a 10-fold reduction in the number of cells required compared with traditional multistep lectin staining methods. Interestingly, we used this one-step lectin array coupled with dimension reduction algorithms in a proof-of-concept application for discrimination among different tumor and immune cell populations. Moreover, this procedure was also able to unveil T-, B-, and myeloid cell subclusters exhibiting differential glycophenotypes. Thus, we report a rapid and versatile lectin cytometry method to simultaneously detect a particular repertoire of surface glycans on living cells that can be easily implemented in different laboratories and core facilities.
Collapse
|
2
|
Balañá ME, Labriola L, Salatino M, Movsichoff F, Peters G, Charreau EH, Elizalde PV. Correction: Activation of ErbB-2 via a hierarchical interaction between ErbB-2 and type I insulin-like growth factor receptor in mammary tumor cells. Oncogene 2023; 42:3087. [PMID: 37684409 DOI: 10.1038/s41388-023-02821-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
|
3
|
Bannoud N, García PA, Gambarte-Tudela J, Sundblad V, Cagnoni AJ, Bach CA, Pérez Saez JM, Blidner AG, Maller SM, Mariño KV, Salatino M, Cerliani JP, Rabinovich GA, Croci DO. Untangling Galectin-Mediated Circuits that Control Hypoxia-Driven Angiogenesis. Methods Mol Biol 2022; 2442:635-653. [PMID: 35320550 DOI: 10.1007/978-1-0716-2055-7_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
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 β-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.
Collapse
|
4
|
Sequeira GR, Sahores A, Dalotto-Moreno T, Perrotta RM, Pataccini G, Vanzulli SI, Polo ML, Radisky DC, Sartorius CA, Novaro V, Lamb CA, Rabinovich GA, Salatino M, Lanari C. Enhanced Antitumor Immunity via Endocrine Therapy Prevents Mammary Tumor Relapse and Increases Immune Checkpoint Blockade Sensitivity. Cancer Res 2021; 81:1375-1387. [PMID: 33268529 DOI: 10.1158/0008-5472.can-20-1441] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/28/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
The role of active antitumor immunity in hormone receptor-positive (HR+) breast cancer has been historically underlooked. The aim of this study was to determine the contribution of the immune system to antiprogestin-induced tumor growth inhibition using a hormone-dependent breast cancer model. BALB/c-GFP+ bone marrow (BM) cells were transplanted into immunodeficient NSG mice to generate an immunocompetent NSG/BM-GFP+ (NSG-R) mouse model. Treatment with the antiprogestin mifepristone (MFP) inhibited growth of 59-2-HI tumors with similar kinetics in both animal models. Interestingly, MFP treatment reshaped the tumor microenvironment, enhancing the production of proinflammatory cytokines and chemokines. Tumors in MFP-treated immunocompetent mice showed increased infiltration of F4/80+ macrophages, natural killer, and CD8 T cells, displaying a central memory phenotype. Mechanistically, MFP induced immunogenic cell death (ICD) in vivo and in vitro, as depicted by the expression and subcellular localization of the alarmins calreticulin and HMGB-1 and the induction of an ICD gene program. Moreover, MFP-treated tumor cells efficiently activated immature dendritic cells, evidenced by enhanced expression of MHC-II and CD86, and induced a memory T-cell response, attenuating tumor onset and growth after re-challenge. Finally, MFP treatment increased the sensitivity of HR+ 59-2-HI tumor to PD-L1 blockade, suggesting that antiprogestins may improve immunotherapy response rates. These results contribute to a better understanding of the mechanisms underlying the antitumor effect of hormonal treatment and the rational design of therapeutic combinations based on endocrine and immunomodulatory agents in HR+ breast cancer. SIGNIFICANCE: Antiprogestin therapy induces immunogenic tumor cell death in PRA-overexpressing tumors, eliciting an adaptive immune memory response that protects mice from future tumor recurrence and increases sensitivity to PD-L1 blockade. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/5/1375/F1.large.jpg.
Collapse
|
5
|
Girotti MR, Salatino M, Dalotto-Moreno T, Rabinovich GA. Sweetening the hallmarks of cancer: Galectins as multifunctional mediators of tumor progression. J Exp Med 2020; 217:133540. [PMID: 31873723 PMCID: PMC7041721 DOI: 10.1084/jem.20182041] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/14/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
Hanahan and Weinberg have proposed 10 organizing principles that enable growth and metastatic dissemination of cancer cells. These distinctive and complementary capabilities, defined as the "hallmarks of cancer," include the ability of tumor cells and their microenvironment to sustain proliferative signaling, evade growth suppressors, resist cell death, promote replicative immortality, induce angiogenesis, support invasion and metastasis, reprogram energy metabolism, induce genomic instability and inflammation, and trigger evasion of immune responses. These common features are hierarchically regulated through different mechanisms, including those involving glycosylation-dependent programs that influence the biological and clinical impact of each hallmark. Galectins, an evolutionarily conserved family of glycan-binding proteins, have broad influence in tumor progression by rewiring intracellular and extracellular circuits either in cancer or stromal cells, including immune cells, endothelial cells, and fibroblasts. In this review, we dissect the role of galectins in shaping cellular circuitries governing each hallmark of tumors, illustrating relevant examples and highlighting novel opportunities for treating human cancer.
Collapse
|
6
|
Perrotta RM, Dalotto-Moreno T, Cagnoni A, Mariño K, Rabinovich G, Salatino M. Abstract 1840: Glycosylation of HER2+ human breast cancer cells control sensitivity to trastuzumab. Cancer Res 2018. [DOI: 10.1158/1538-7445.am2018-1840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Galectins decode glycan-containing information in a number of cell receptors adjusting signaling thresholds and modulating cellular functions. Upon specific binding to cell surface glycans these lectins control receptor clustering, endocytosis and signaling, influencing many physiological and pathological conditions. In particular, galectin-1 (Gal1), a 14kD prototype galectin, binds to terminal N-acetyllactosamine residues on glycosylated proteins in the absence of α2-6 sialic acid capping (Gal1 permissive glycophenotype). In early studies, our group demonstrated that tumors usurp the Gal1 pathway to evade immune surveillance and to promote aberrant angiogenesis, influencing sensitivity to anti-VEGF therapy. Here we aim to explore the glycosylation signature of HER2+ breast cancer cells and to investigate the effects of tumor-derived Gal1 on RTK activation and resistance to trastuzumab (TZ). We first selected three HER2+ human breast cancer cell lines with different response to TZ: JIMT-1 (resistant-TZR), BT-474 and SK-BR-3 (sensitive-TZS). To identify specific glycan structures, we used a panel of biotinylated plant lectins with different carbohydrate specificities. We found that TZS cell lines exhibited a Gal-1 restrictive glycophenotype characterized by high α2,6 sialic acid capping. Profiling of N-type glycans (WAX-HPLC) confirmed that TZSs cell lines exhibit higher frequency of α2,6 sialic acid residues compared to TZR JIMT-1. This effect was further substantiated by RT-PCR analysis of specific glycosyltransferases responsible of Gal1 ligands biosynthesis. These results were supported by analysis of raw data from public databases arrays (GSE62327) showing that patients who presented complete response to TZ exhibited higher levels of ST6GAL1, a glycosyltransferase responsible for α2-6 sialic acid capping. In accordance with the glycophenotype, TZR cell line bound and expressed higher levels of Gal1 when compared to TZS cell lines by Western blot, RT-PCR and ELISA of conditioned medium, suggesting a positive autocrine loop that could modulate cell behavior. Moreover, in silico analysis of raw data from the Long HER Study (GSE44272) revealed that patients with poor response to TZ expressed higher levels of Gal1 mRNA than long-term responders, reinforcing our hypothesis from a clinical standpoint. Finally, in order to elucidate Gal1 implications in TZ resistance, we knocked down Gal1 in JIMT-1 cell line using shRNA strategies. Remarkably, absence of Gal1 expression sensitized JIMT-1 cells to TZ-inhibition in vitro, and the resistant phenotype was further reestablished by addition of human recombinant Gal1. In summary, our study suggests that individual HER2+ human breast cancer cells display particular “glycosylation signatures” which, in association with Gal1 expression pattern, may control resistance to anti-HER2 targeted therapy and may predict breast cancer clinical outcome.
Citation Format: Ramiro M. Perrotta, Tomás Dalotto-Moreno, Alejandro Cagnoni, Karina Mariño, Gabriel Rabinovich, Mariana Salatino. Glycosylation of HER2+ human breast cancer cells control sensitivity to trastuzumab [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1840.
Collapse
|
7
|
Perrotta RM, Dalotto-Moreno T, Cagnoni A, Mariño KV, Rabinovich GA, Salatino M. Abstract P4-03-08: Withdrawn. Cancer Res 2018. [DOI: 10.1158/1538-7445.sabcs17-p4-03-08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
This abstract was withdrawn by the authors.
Collapse
|
8
|
Abstract
The clinical efficacy of therapies targeting the PD-1/PD-L1 pathway is still limited. In this issue of Cancer Cell, Li and colleagues identify a PD-L1 glycosylation-based mechanism in triple-negative breast cancer that fosters immunosuppression by enhancing interactions with PD-1. Targeting glycosylated PD-L1 with a drug-conjugated antibody opens new avenues for treatment.
Collapse
|
9
|
Dalotto Moreno T, Cerliani JP, Croci DO, Mendez-Huergo SP, Moses F, Rabinovich GA, Salatino M. Abstract 465: Progestin-driven regulatory T cells directly promote an aggressive and metastatic phenotype in triple-negative breast cancer. Cancer Res 2015. [DOI: 10.1158/1538-7445.am2015-465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
The immune system plays key roles in the recognition and elimination of most tumors. Progesterone (Pg) can shape the immune response favoring a tolerogenic rather than a pro-inflammatory adaptive response. As hormone replacement therapies, supplementation and hormone-based contraceptives have been associated with an increase of malignant breast neoplasia we decided to investigate how progestins can regulate different key immune cell populations in the tumor microenvironment and their influence to tumor progression. To address this issue we used the highly metastatic, triple-negative 4T1 breast tumor. Balb/c mice treated with Pg or its synthetic analog, medroxyprogesterone acetate, showed an increased frequency of Foxp3+ regulatory T cells (Tregs) in draining lymph nodes (DLN) and an impaired antitumor response elicited by a decreased production of IL-17 and IFN-γ by CD4+ T cells (p<0,01). Tregs in DLN of progestin-treated animals showed increased expression of CD44 and CTLA-4. CD44+ Tregs were more suppressive in vitro than their CD44- counterpart. Concordantly, progestin-treated tumor-bearing mice showed a higher frequency of CD44+ Tregs and PD1+TIM3+ ‘exhausted’ CD8 T cells in the tumor (p<0,05). Both progestins favored in vitro Treg stable differentiation and expansion which resulted in a more robust suppression activity (p<0,01). Progestin-induced Treg differentiation could not be inhibited by the nuclear progesterone receptor (nPR) antagonist, Zk230211. Interestingly, T cells did not express detectable levels of nPR by qPCR but did express membrane progesterone receptor alfa, gamma and progesterone receptor membrane component type 1. In the presence of progestins, in vitro differentiated Tregs expressed higher levels of RANKL, a protein involved in breast stem cell maintenance and malignant transformation of tumors. When cocultured, ex vivo sorted- or in vitro differentiated Tregs increased 4T1 cells invasiveness both in vitro and in vivo following the downregulation of E-cadherin and increased expression of Snail (p = 0,05). Concordantly we observed a stable expansion of the CD44+ stem cell-like population when cocultured with progestin-iTregs. The CD44+ 4T1 population formed tumor spheres, exhibited an EMT molecular signature and had augmented tumorigenicity and a metastatic phenotype. Although primary tumor volume was not altered, the number of lung metastases was notably increased in animals treated with progestins (p<0,01). Additonally, overall survival of mice was decreased when injected with 4T1 cells cocultured with progestin-induced Tregs compared with 4T1 cocultured with control Tregs. Our findings highlight the relevance of progestins in modulating immunoregulatory checkpoints and harnessing immunosurveillance in the tumor microenvironment and suggest a mechanism through which Tregs could directly promote a metastatic and aggressive phenotype on tumor cells.
Citation Format: Tomas Dalotto Moreno, Juan Pablo Cerliani, Diego Omar Croci, Santiago Patricio Mendez-Huergo, Florencia Moses, Gabriel Adrian Rabinovich, Mariana Salatino. Progestin-driven regulatory T cells directly promote an aggressive and metastatic phenotype in triple-negative breast cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 465. doi:10.1158/1538-7445.AM2015-465
Collapse
|
10
|
Blidner AG, Salatino M, Mascanfroni ID, Diament MJ, Bal de Kier Joffé E, Jasnis MA, Klein SM, Rabinovich GA. Differential Response of Myeloid-Derived Suppressor Cells to the Nonsteroidal Anti-Inflammatory Agent Indomethacin in Tumor-Associated and Tumor-Free Microenvironments. THE JOURNAL OF IMMUNOLOGY 2015; 194:3452-62. [DOI: 10.4049/jimmunol.1401144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
11
|
Rutkowski MR, Stephen TL, Svoronos N, Allegrezza MJ, Tesone AJ, Perales-Puchalt A, Brencicova E, Escovar-Fadul X, Nguyen JM, Cadungog MG, Zhang R, Salatino M, Tchou J, Rabinovich GA, Conejo-Garcia JR. Microbially driven TLR5-dependent signaling governs distal malignant progression through tumor-promoting inflammation. Cancer Cell 2015; 27:27-40. [PMID: 25533336 PMCID: PMC4293269 DOI: 10.1016/j.ccell.2014.11.009] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/29/2014] [Accepted: 11/08/2014] [Indexed: 02/07/2023]
Abstract
The dominant TLR5(R392X) polymorphism abrogates flagellin responses in >7% of humans. We report that TLR5-dependent commensal bacteria drive malignant progression at extramucosal locations by increasing systemic IL-6, which drives mobilization of myeloid-derived suppressor cells (MDSCs). Mechanistically, expanded granulocytic MDSCs cause γδ lymphocytes in TLR5-responsive tumors to secrete galectin-1, dampening antitumor immunity and accelerating malignant progression. In contrast, IL-17 is consistently upregulated in TLR5-unresponsive tumor-bearing mice but only accelerates malignant progression in IL-6-unresponsive tumors. Importantly, depletion of commensal bacteria abrogates TLR5-dependent differences in tumor growth. Contrasting differences in inflammatory cytokines and malignant evolution are recapitulated in TLR5-responsive/unresponsive ovarian and breast cancer patients. Therefore, inflammation, antitumor immunity, and the clinical outcome of cancer patients are influenced by a common TLR5 polymorphism.
Collapse
|
12
|
Salatino M, Croci DO, Laderach DJ, Compagno D, Gentilini L, Dalotto-Moreno T, Dergan-Dylon LS, Méndez-Huergo SP, Toscano MA, Cerliani JP, Rabinovich GA. Regulation of galectins by hypoxia and their relevance in angiogenesis: strategies and methods. Methods Mol Biol 2015; 1207:293-304. [PMID: 25253148 DOI: 10.1007/978-1-4939-1396-1_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
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.
Collapse
|
13
|
Salatino M, Dalotto T, Croci DO, Mendez Huergo SP, Dergan Dylon SL, Cerliani JP, Toscano MA, Rabinovich GA. Abstract 449: Progesterone-induced immunosuppression thwarts immunosurveillance to tumors and promotes lung metastasis in a breast cancer model. Immunology 2014. [DOI: 10.1158/1538-7445.am2013-449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
14
|
Salatino M, Dalotto-Moreno T, Rabinovich GA. Thwarting galectin-induced immunosuppression in breast cancer. Oncoimmunology 2014; 2:e24077. [PMID: 23762796 PMCID: PMC3667902 DOI: 10.4161/onci.24077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 02/21/2013] [Indexed: 12/14/2022] Open
Abstract
Novel therapeutic strategies are needed to counteract breast cancer-associated immunosuppression. Silencing the expression of galectin-1 in a breast carcinoma model inhibited tumor growth and prevented lung metastasis by reducing the frequency and immunosuppressive activity of CD4+CD25+ FOXP3+ regulatory T cells.
Collapse
|
15
|
Croci DO, Cerliani JP, Dalotto-Moreno T, Méndez-Huergo SP, Mascanfroni ID, Dergan-Dylon S, Toscano MA, Caramelo JJ, García-Vallejo JJ, Ouyang J, Mesri EA, Junttila MR, Bais C, Shipp MA, Salatino M, Rabinovich GA. Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors. Cell 2014; 156:744-58. [PMID: 24529377 DOI: 10.1016/j.cell.2014.01.043] [Citation(s) in RCA: 400] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 11/11/2013] [Accepted: 01/21/2014] [Indexed: 02/07/2023]
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 β1-6GlcNAc 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.
Collapse
|
16
|
Spallanzani RG, Dalotto-Moreno T, Raffo Iraolagoitia XL, Ziblat A, Domaica CI, Avila DE, Rossi LE, Fuertes MB, Battistone MA, Rabinovich GA, Salatino M, Zwirner NW. Expansion of CD11b(+)Ly6G (+)Ly6C (int) cells driven by medroxyprogesterone acetate in mice bearing breast tumors restrains NK cell effector functions. Cancer Immunol Immunother 2013; 62:1781-95. [PMID: 24114144 PMCID: PMC11028897 DOI: 10.1007/s00262-013-1483-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/01/2013] [Indexed: 12/13/2022]
Abstract
The progesterone analog medroxyprogesterone acetate (MPA) is widely used as a hormone replacement therapy in postmenopausal women and as contraceptive. However, prolonged administration of MPA is associated with increased incidence of breast cancer through ill-defined mechanisms. Here, we explored whether exposure to MPA during mammary tumor growth affects myeloid-derived suppressor cells (MDSCs; CD11b(+)Gr-1(+), mostly CD11b(+)Ly6G(+)Ly6C(int) and CD11b(+)Ly6G(-)Ly6C(high) cells) and natural killer (NK) cells, potentially restraining tumor immunosurveillance. We used the highly metastatic 4T1 breast tumor (which does not express the classical progesterone receptor and expands MDSCs) to challenge BALB/c mice in the absence or in the presence of MPA. We observed that MPA promoted the accumulation of NK cells in spleens of tumor-bearing mice, but with reduced degranulation ability and in vivo cytotoxic activity. Simultaneously, MPA induced a preferential expansion of CD11b(+)Ly6G(+)Ly6C(int) cells in spleen and bone marrow of 4T1 tumor-bearing mice. In vitro, MPA promoted nuclear mobilization of the glucocorticoid receptor (GR) in 4T1 cells and endowed these cells with the ability to promote a preferential differentiation of bone marrow cells into CD11b(+)Ly6G(+)Ly6C(int) cells that displayed suppressive activity on NK cell degranulation. Sorted CD11b(+)Gr-1(+) cells from MPA-treated tumor-bearing mice exhibited higher suppressive activity on NK cell degranulation than CD11b(+)Gr-1(+) cells from vehicle-treated tumor-bearing mice. Thus, MPA, acting through the GR, endows tumor cells with an enhanced capacity to expand CD11b(+)Ly6G(+)Ly6C(int) cells that subsequently display a stronger suppression of NK cell-mediated anti-tumor immunity. Our results describe an alternative mechanism by which MPA may affect immunosurveillance and have potential implication in breast cancer incidence.
Collapse
MESH Headings
- Animals
- Antigens, Ly/immunology
- Antigens, Ly/metabolism
- Antineoplastic Agents, Hormonal/pharmacology
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- CD11b Antigen/immunology
- CD11b Antigen/metabolism
- Cell Differentiation
- Cell Proliferation
- Cytotoxicity, Immunologic
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Humans
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Medroxyprogesterone Acetate/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Myeloid Cells/drug effects
- Myeloid Cells/immunology
- Myeloid Cells/metabolism
- Receptors, Glucocorticoid/metabolism
- STAT3 Transcription Factor/metabolism
- Tumor Cells, Cultured
Collapse
|
17
|
Croci D, Cerliani J, D Alotto T, Mascanfroni I, Salatino M, Mendez-Huergo S, Rabinovich G. Targeted disruption of lectin-glycan interactions promotes remodeling of tumor vascular networks and restores immune function. (P2173). THE JOURNAL OF IMMUNOLOGY 2013. [DOI: 10.4049/jimmunol.190.supp.170.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The mechanisms linking tumor neovascularization and immunity are poorly understood. We previously demonstrated an essential role of galectin-1 (Gal-1) in tumor-immune escape and angiogenesis. The present study was conducted to elucidate whether Gal1-glycan lattices can link tumor angiogenesis to immunity. We examined the glycophenotype of endothelial cells (ECs) in resting, proliferative, tolerogenic or inflammatory conditions. ECs exposed to tolerogenic or proliferative microenvironments exhibited a substantial up-regulation of cell surface glycans critical for Gal-1 binding (p<0.01). In vivo disruption of Gal1-glycan lattices in B16 melanomas attenuated hypoxia-driven angiogenesis, while promoting vascular remodeling (p<0.01) in tumors treated with anti-Gal1 blocking mAb. Moreover, anti-Gal1 treated tumors showed a significant reduction in tumor growth (p<0.01) and evoked a T-cell specific immune responses, as shown by increased infiltration of CD8+ T-cell (p<0.05), increased proliferation of tumor-specific CD4+ T-cells (p<0.01) and augmented IFN-γ and IL-17 (p<0.05) production. Moreover, tumor-draining LN of mice receiving anti-Gal-1 mAb showed lower frequency of FoxP3+ Treg cells (p<0.05) and lower IL-10 secretion (p<0.05). Hence, disruption of lectin-glycan lattices, not only evokes an unleashed anti tumor immune response, but also favors remodeling of tumor vascular networks, highlighting the versatility of endogenous lectins during cancer progression.
Collapse
|
18
|
Zwirner N, Spallanzani R, Dalotto T, Rossi L, Avila D, Ziblat A, Domaica C, Fuertes M, Raffo Iraolagoitia X, Rabinovich G, Salatino M. Medroxiprogesterone acetate drives expansion of CD11b+Gr1high myeloid-derived suppressor cells which suppress NK cell effector functions in tumor-bearing mice (P2059). THE JOURNAL OF IMMUNOLOGY 2013. [DOI: 10.4049/jimmunol.190.supp.53.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Abstract
Hormone replacement therapy with the progesterone analogue medroxyprogesterone acetate (MPA) is widely used in postmenopausal women for the treatment of endometrial conditions, and as a contraceptive. However, prolonged administration of MPA is associated with increased incidence of breast cancer through ill-defined mechanisms. Our aim was to explore if MPA threats immunosurveillance to tumors by effecting myeloid-derived suppressor (MDSCs; CD11b+Gr1+) and NK cells in mammary tumor-bearing mice. We used the highly metastatic 4T1 breast tumor which does not express classical progesterone or glucocorticoid receptors. MPA did not affect primary tumor growth in 4T1-tumor bearing mice but promoted lung metastasis burden. This effect was accompanied by expansion of splenic CD11b+Gr1+ cells (mostly CD11b+Gr1high cells). Sorted CD11b+Gr1+ cells from MPA-treated tumor bearing mice showed a more pronounced inhibitory activity of NK cell degranulation in response to YAC-1 cells and IFN-γ production in response to cytokines than those isolated from untreated tumor-bearing mice. Also, MPA significantly increased the percentage of spleen NK cells in tumor-bearing mice with similar lung infiltration of CD11b+Gr1+ and NK cells as compared to untreated tumor-bearing mice. We conclude that in breast cancer-bearing hosts MPA promotes the accumulation of CD11b+Gr1+ which suppress NK-cell mediated anti-tumor activity potentially contributing to tumor progression and metastasis.
Collapse
|
19
|
Dalotto-Moreno T, Croci DO, Cerliani JP, Martinez-Allo VC, Dergan-Dylon S, Méndez-Huergo SP, Stupirski JC, Mazal D, Osinaga E, Toscano MA, Sundblad V, Rabinovich GA, Salatino M. Targeting galectin-1 overcomes breast cancer-associated immunosuppression and prevents metastatic disease. Cancer Res 2012. [PMID: 23204230 DOI: 10.1158/0008-5472.can-12-2418] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
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.
Collapse
|
20
|
Croci DO, Salatino M, Rubinstein N, Cerliani JP, Cavallin LE, Leung HJ, Ouyang J, Ilarregui JM, Toscano MA, Domaica CI, Croci MC, Shipp MA, Mesri EA, Albini A, Rabinovich GA. Disrupting galectin-1 interactions with N-glycans suppresses hypoxia-driven angiogenesis and tumorigenesis in Kaposi's sarcoma. ACTA ACUST UNITED AC 2012; 209:1985-2000. [PMID: 23027923 PMCID: PMC3478924 DOI: 10.1084/jem.20111665] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Disrupting Gal-1 interactions with N-glycans prevents hypoxia-driven angiogenesis to suppress tumorigenesis of Kaposi’s sarcoma 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.
Collapse
|
21
|
Moreno TD, Croci DO, Cerliani JP, Dylon SLD, Mascanfroni I, Osinaga E, Shipp MA, Rabinovich GA, Salatino M. Abstract 3547: Disruption of Galectin1-glycan interaction impairs tumor growth and metastasis in breast cancer by disarming the immunosuppressive capacity of regulatory T cells. Cancer Res 2012. [DOI: 10.1158/1538-7445.am2012-3547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Galectin-1 (Gal1) is an endogenous lectin that emerged as a key regulator of immune cell homeostasis. By binding to surface glycoconjugates, Gal1 modulates diverse physiological processes and also shapes the immune response during cancer progression to a suppressive phenotype. For that reason Gal1 has become a molecule with a profound impact in the tumor microenvironment biology. In the present work we investigated whether tumor Gal1 expression contributes to immunosuppression, tumor growth and metastasis in breast cancer in order to validate the Gal-1-glycan axis as a novel therapeutic target in these tumors. We used the highly metastatic mouse mammary tumor model 4T1 which expresses high levels of Gal1. We generated Gal1-deficient 4T1 cell line (4T1 KD) using a retrovirus encoding a shRNA specific for mouse Gal1. After injecting Balb/c mice with either 4T1 WT or KD cells we studied tumor growth, metastasis formation and analyzed tumor-associated immune compartments. Silencing of Gal-1 induced a marked reduction in both tumor growth and the number of lung metastases. This effect was abrogated if a 4T1 WT tumor was inoculated in the contralateral flank of these mice, suggesting that the antitumor effect involves modulation of the immune system. In regard to the latter we observed that 4T1 WT tumor-bearing mice exhibited a higher frequency of CD4+CD25+Foxp3+ regulatory T cells (Tregs) in the lungs, spleen, tumor draining lymph nodes (TDLN) and tumor microenvironment. Remarkably, knocking down Gal1 reverted the systemic immune tolerance that characterizes tumor progression, and decreased the frequency and immunosuppressive function of Tregs as evidenced by a suppression assay and expression of Tregs tolerogenic molecular markers as Foxp3, TGF-β and LAT. In this sense, mice bearing Gal-1 KD tumors were able to reject an allogeneic B16 tumor similarly to tumor free mice. In addition, antigen-specific proliferation of lymphocytes purified from 4T1 KD-bearing mice was increased when reestimulated ex vivo. Concomitantly, blockade of tumor Gal1 was accompanied by a reduction of the IL-10/IFNγ cytokines ratio. Finally, therapeutic administration of a Gal1 neutralizing mAb (F8.G7) to 4T1 WT tumor bearing mice not only induced a reduction in tumor growth and lung metastasis formation but also reverted tumor associated immunosuppression. These results are strengthened by the observation that in human breast cancer biopsies Gal1 expression correlates with tumor grade (Bloom-Richardson histopathological grade I vs II-III). In conclusion our results indicate that disruption of Gal1 impairs tumor growth and metastasis via mechanisms involving reversal of systemic immunosuppression and further validate the Gal1-glycan axis as an attractive target in the therapeutic treatment of metastasic breast cancer.
Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3547. doi:1538-7445.AM2012-3547
Collapse
|
22
|
Zacarías Fluck MF, Hess L, Salatino M, Croci DO, Stupirski JC, Di Masso RJ, Roggero E, Rabinovich GA, Scharovsky OG. The aggressiveness of murine lymphomas selected in vivo by growth rate correlates with galectin-1 expression and response to cyclophosphamide. Cancer Immunol Immunother 2012; 61:469-80. [PMID: 21947259 PMCID: PMC11029055 DOI: 10.1007/s00262-011-1114-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/07/2011] [Indexed: 12/19/2022]
Abstract
Although lymphomas account for almost half of blood-derived cancers that are diagnosed each year, the causes of new cases are poorly understood, as reflected by the relatively few risk factors established. Galectin-1, an immunoregulatory ß-galactoside-binding protein, has been widely associated with tumor-immune escape. The aim of the present work was to study the relationship between tumor growth rate, aggressiveness, and response to cyclophosphamide (Cy) therapy with regard to Gal-1 expression in murine T-cell lymphoma models. By means of a disruptive selection process for tumor growth rate, we generated two lymphoma variants from a parental T-cell lymphoma, which have unique characteristics in terms of tumor growth rate, spontaneous regression, metastatic capacity, Gal-1 expression and sensitivity to Cy therapy. Here, we show that Gal-1 expression strongly correlates with tumor growth rate, metastatic capacity and response to single-dose Cy therapy in T-cell lymphoma models; this association might have important consequences for evaluating prognosis and treatments of this type of tumors.
Collapse
|
23
|
O. Croci D, Salatino M. Tumor Immune Escape Mechanisms that Operate During Metastasis. Curr Pharm Biotechnol 2011; 12:1923-36. [DOI: 10.2174/138920111798376987] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 05/24/2010] [Indexed: 11/22/2022]
|
24
|
Maldonado CA, Sundblad V, Salatino M, Elia J, García LN, Leimgruber C, Croci DO, Rabinovich GA. Cell-type specific regulation of galectin-3 expression by glucocorticoids in lung Clara cells and macrophages. Histol Histopathol 2011; 26:747-59. [PMID: 21472689 DOI: 10.14670/hh-26.747] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bronchiolar Clara cells are integral components of lung homeostasis, predominantly distributed in distal airways. In addition to the 16 kDa Clara cell protein, a major secretory product with anti-inflammatory effects, rat Clara cells express the glycan-binding protein galectin-3 and secrete it into the airways. Given the essential role of galectin-3 in the control of inflammation and the well-established function of glucocorticoids (GCs) in lung physiology, here we investigated whether galectin-3 is a target of the regulatory effects of GCs. Adult male rats were subjected to bilateral adrenalectomy and the lungs were processed for light and transmission electron microscopy, immunoelectron microscopy and Western blot analysis. Profound changes in bronchiolar Clara cells and macrophage morphology could be observed by electron microscopy after adrenalectomy. While specific galectin-3 staining was detected in the nucleus and cytoplasm of Clara cells and macrophages from control animals, cytoplasmic galectin-3 expression was dramatically reduced after adrenalectomy in both cell types. This effect was cell-specific as it did not affect expression of this lectin in ciliated cells. After dexamethasone treatment, galectin-3 expression increased significantly in the nucleus and cytoplasm of macrophages and Clara cells. Western blot analysis showed a clear decrease in galectin-3 expression in ADX animals, which was recovered after a 7-day treatment with dexamethasone. In peritoneal macrophages, galectin-3 expression was also dependent on the effects of GCs both in vivo and in vitro. Our results identify a cell type-specific control of galectin-3 synthesis by GCs in lung bronchiolar Clara cells and interstitial macrophages, which may provide an alternative mechanism by which GCs contribute to modulate the inflammatory response.
Collapse
|
25
|
Carlini MJ, Vazquez P, Diament M, Salatino M, Rabinovich GA, Bal de Kier Joffe ED, Puricelli LI. Abstract 400: Modulation of the metastatic microenvironment by soluble factors released by lung adenocarcinoma cells. Cancer Res 2011. [DOI: 10.1158/1538-7445.am2011-400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Soluble factors from primary tumors can modify target organs for metastasis through mobilization of bone marrow derived cells (BMDCs), which are recruited to tumors and to metastatic target organs and facilitate tumor growth and metastasis. Here we aimed to study early cellular and molecular events occurring in the lung, the metastatic target organ of the murine lung adenocarcinoma cell line LP07.
We assessed by flow cytometry the numbers of BMDC positive for VEGFR1, SCF, CD34 or CXCR4 and the numbers of myeloid suppressor cells (CD11b+/Gr1+) and regulatory T cells (CD4+/CD25+/FOXP3+) in single cell lung suspensions obtained from 15-day tumor-bearing and control mice. Control and tumor-conditioned lungs of inoculated mice showed comparable amounts of these cell populations. To assess whether the primary tumor stimulates subsequent formation of macroscopic metastasis, mice bearing an intraperitoneal millipore chamber -that allows the diffusion of soluble molecules but not cell extravasation- loaded with LP07 cells or medium, were challenged with a LP07 intravenous inoculum at day 15. The presence of LP07 loaded chambers decreased the number of lung metastases three weeks after the inoculum compared to control mice (p≪0.05). In a different approach, mice were daily injected for 15 days with LP07 conditioned medium or control medium, and then challenged with a LP07 intravenous inoculum. When evaluated at day 21 post LP07 cell inoculation, the number of metastatic lung nodules was significantly lower (p≪0.05) in the group treated with LP07 conditioned medium. Besides, when we assessed matrix metalloproteinase (MMP) activity in lung homogenates from 15 day-tumor bearing and control mice, we found a significant increase in MMP-9 activity (p≪0.05) as assessed by semi-quantitative zymography in lungs from tumor-bearing mice.In conclusion, in the LP07 lung adenocarcinoma model, tumor cells secrete soluble factors that induce certain changes in the metastatic target organ, as the increase of MMP-9 activity. Moreover, in this experimental metastatic approach, a decrease in metastasis development was observed; the mechanisms underlying this phenomenon deserve further investigation.
Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 400. doi:10.1158/1538-7445.AM2011-400
Collapse
|